WO1989006546A1 - Conjugation of polymer to colony stimulating factor-1 - Google Patents

Conjugation of polymer to colony stimulating factor-1 Download PDF

Info

Publication number
WO1989006546A1
WO1989006546A1 PCT/US1989/000270 US8900270W WO8906546A1 WO 1989006546 A1 WO1989006546 A1 WO 1989006546A1 US 8900270 W US8900270 W US 8900270W WO 8906546 A1 WO8906546 A1 WO 8906546A1
Authority
WO
WIPO (PCT)
Prior art keywords
lcsf
protein
csf
scsf
tyr
Prior art date
Application number
PCT/US1989/000270
Other languages
French (fr)
Inventor
Paula J. Shadle
Kirston E. Koths
Margaret Moreland
Nandini Katre
Walter J. Laird
Lois Aldwin
Danute E. Nitecki
John D. Young
Original Assignee
Cetus Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cetus Corporation filed Critical Cetus Corporation
Priority to DE68913520T priority Critical patent/DE68913520T2/en
Priority to AT89902670T priority patent/ATE102050T1/en
Publication of WO1989006546A1 publication Critical patent/WO1989006546A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/53Colony-stimulating factor [CSF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/58Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. poly[meth]acrylate, polyacrylamide, polystyrene, polyvinylpyrrolidone, polyvinylalcohol or polystyrene sulfonic acid resin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S930/00Peptide or protein sequence
    • Y10S930/01Peptide or protein sequence
    • Y10S930/14Lymphokine; related peptides
    • Y10S930/145Colony stimulating factor
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S930/00Peptide or protein sequence
    • Y10S930/01Peptide or protein sequence
    • Y10S930/30Signal or leader sequence

Definitions

  • This invention relates to a chemical modification of biologically active colony stimulating factor- 1 (CSF-1) that alters the chemical and/or physiological properties of this protein. More specifically, this invention relates to selective conjugation of CSF-1 to polymers to increase the circulating half-life of the protein in mammals.
  • CSF-1 biologically active colony stimulating factor-1
  • Colony stimulating factor- 1 (also known as M-CSF) is one of several proteins that are capable of stimulating colony formation by bone marrow cells plated in semisolid culture medium. CSF-1 is distinguished from other colony stimulating factors by its ability to stimulate the formation of predominantly macrophage colonies. Other CSFs stimulate the production of colonies that consist of neutrophilic granulocytes and macrophages, exclusively neutrophilic granulocytes, or neutrophilic and eosinophilic granulocytes and macrophages. A review of these CSFs has been published by Dexter, T.M., Nature (1984) 202:746, and by Vadas, M.A.. J. Immunol (1983) 130:793. There is currendy no routine in vivo assay that is known to be specific for CSF-1 activity.
  • CSF-1 has been purified from native sources (see, e.g., Csejtey et al., Biochem. Biophvs. Res. Comm. (1986) 12&238 and PCT publication No. WO 86/04587 published August 14, 1986 regarding immunoaffinity chromatography of native CSF-1 to enable partial amino acid determinations).
  • CSF-1 has also been produced from recombinant DNA using two apparendy related cDNA clones: (1) a "short” form that encodes a monomeric protein of 224 amino acids preceded by a 32-amino acid signal sequence (Kawasaki et al., Science (1985) 220:292-296); and (2) a "long” form, encoding a monomeric protein of 522 amino acids, also preceded by the 32-amino acid signal sequence.
  • the long form has been cloned and expressed by two groups, as disclosed in European Patent Publication No. 0272779 published June 29, 1988 and Ladner et al. EMBO J.
  • the long and short forms of the CSF-1-encoding DNA appear to arise from a variable splice junction at the upstream portion of exon 6 of the genomic CSF-1-encoding DNA.
  • CSF-1 When CSF-1 is expressed in certain eucaryotic cells from either the long or short cDNA forms, it is secreted as a dimeric glycoprotein and appears to be variably processed at the C-terminus and/or variably glycosylated. Consequently, CSF-1 proteins of varying molecular weights are found when the reduced monomeric form is subjected to Western analysis.
  • amino acid sequences of the long and short forms are identical in the first 149 amino acids at the N-terminus after signal peptide cleavage, and diverge thereafter as a result of the insertion in the longer clone of an additional 894 bp fragment (encoding 298 additional amino acids) before the codon encoding amino acid 150. Therefore, both the shorter and longer forms of the gene encode regions of identical sequence at the C-terminus, as well as at the N-terminus.
  • Biologically active protein has been recovered when truncated cDNAs encoding only the first 145 or 147 amino acids of the mature short form (European -Patent Publication No.0261592 published March 30, 1988; Cerretti et al. supra or the first 190 or 221 amino acids of the mature longer form, are expressed in eucaryotic cells.
  • Recombinant CSF-1 was expressed in I £c_U by modifying a short clone cDNA originally described by Kawasaki et al., Science (1985) 230:291 to code for proteins that contained 1) the native N-terminus and a C- terminus at amino acid 150 of the mature protein, and 2) a truncation to delete the first two amino acids at the N-terminus and a C-terminus at amino acid 150 of the mature protein. These proteins were purified and refolded to form homodimers and were found to have apparent molecular weights on size- exclusion high performance liquid chromatography (HPLC) of about 43,000 and 40,000 daltons, respectively.
  • HPLC size- exclusion high performance liquid chromatography
  • CSF-1 proteins modified so that the C- terminus of the expressed protein is amino acid 150 or 158 and so that up to three amino acids at the N-terminus are deleted have also been prepared.
  • Small proteins (less than about 70 kd) often have a relatively short half-life in blood after intravenous injection. Rapid clearance of drugs from circulation often reduces their efficacy. It is often desirable to increase the half-life of a circulating polypeptide so that smaller amounts of the polypeptide or less frequent injections might be administered, while retaining the desired therapeutic effect
  • Modifications of the CSF-1 protein that might alter its half-life in vivo, reduce its immunogenicity, or reduce or eliminate aggregation of the protein that might occur when it is introduced in vivo would be desirable.
  • Such modifications include the modification of proteins with substantially straight chain polymers such as polyethylene glycol (PEG), polypropylene glycol (PPG), dextran, or polyvinyl alcohol.
  • PEG polyethylene glycol
  • PPG polypropylene glycol
  • dextran or polyvinyl alcohol
  • U.S. Patent No. 4,261,973 describes conjugation of immunogenic allergen molecules with non-immunogenic water- soluble polymers such as PEG or polyvinyl alcohol to reduce the immunogenicity of the allergen.
  • U.S. Patent No. 4,301,144 describes conjugation of hemoglobin to PEG, PPG, a copolymer of ethylene glycol with propylene glycol, or ethers, esters or dehydrated products of such polymers to increase the oxygen-carrying ability of the hemoglobin molecule.
  • Patent No.4,609,546 discloses that conjugation of a polypeptide or glycoprotein such as a colony stimulating factor to a polyoxyethylene-polyoxypropylene copolymer may increase the duration of its physiological activity.
  • the only proteins that have been tested in this fashion are enzymes or native interferon, which are readily water-soluble.
  • PCT WO 86/04145 published July 17, 1986 discloses PEG modification of antibodies to decrease binding to Fc receptors.
  • U.S. Patent No. 4,179,337 discloses conjugation of water-soluble polypeptides such as enzymes and insulin to PEG or PPG to reduce the immunogenicity of the polypeptides while retaining a substantial proportion of their desired physiological activities.
  • EP 154,316 published September 11, 1985 to Takeda Chemical Industries, Ltd., discloses and claims chemically modified lymphokines such as IL-2 containing PEG bonded directly to at least one primary amino group of the lymphokine.
  • Katre et al. Proc. Natl. Acad. Sci. (1987) 8_4: 1487 discloses modification of IL-2 with PEG.
  • Patents and patent publications that disclose use of polyvinyl alcohol (PVA) in protein conjugation reactions include U.S. Patent Nos. 4,296,097 and 4,430,260, relating to conjugation of benzylpenicilliri and PVA, U.S. Patent No.
  • Articles relating to conjugates of proteins and PVA include Sabet et al., Indian J. Chem.. Sec. A (1984) 23A(5) (disclosing PVA and protein interaction), Wei et al., Immunol. (1984) 5I(4):687-696 (disclosing trimellityl conjugated with PVA), Lee et al., J. Immunol. (1981) 126:414-418 and Hubbard et al., J. Immunol. (1981) 126:407-413 (both disclosing DNP conjugated to PVA), Lee et al., Int. Arch. Allergy Appl. Immunol.
  • the present invention provides for modifying colony stimulating factor- 1 to increase its half-life in vivo, while also retaining useful biological activity.
  • the modified CSF-1 may be used to stimulate cells In vivo at much reduced and/or less frequent dosage than the unmodified CSF- 1.
  • the modification may decrease the immunogenicity of the CSF-1 (especially when used in heterologous species, such as use of human CSF-1 for cattle) and/or reduce aggregation of the protein that might occur.
  • the present invention is directed to a biologically active composition having prolonged in vivo half-life in mammals, comprising a protein that stimulates the formation of primarily macrophage colonies in the in vitro colony stimulating factor- 1 assay, which protein is covalendy conjugated to a water-soluble polymer selected from the group consisting of polyethylene glycol or polypropylene glycol homopolymers, polyoxyethylated polyols, and polyvinyl alcohol, wherein said homopolymer is unsubstituted or substituted at one end with an alkyl group.
  • the CSF-1 protein may be conjugated to the polymer via: (1) free amino groups, including lysine residues and the N-terminal amino acid (preferably 1 to 3 sites), (2) carbohydrate moiety(ies) of eukaryote-expressed, glycosylated CSF-1; or (3) free sulfhydryl groups engineered into the CSF-1
  • the polymer is unsubstituted polyethylene glycol (PEG), monomethyl PEG (mPEG), or polyoxyethylated glycerol (POG) that is coupled to the (1) lysine residue(s) of the CSF-1 via an amide or urethane linkage formed from an active ester (preferably the N-hydroxysuc ⁇ nimide or paranitrophenyl ester) of a PEG, mPEG, or POG carboxylic or carbonic acid; (2) carbohydrate moiety(ies) of the CSF-1 via an amine, hydrazine, or hydrazide linkage; or (3) cyst
  • Another aspect of this invention resides in a process for preparing the conjugated protein described above, comprising:
  • the invention in another aspect, relates to a method for prophylactic or therapeutic treatment of infectious diseases or cancer in mammals and to a methods effective for cancer treatment, osteroporesis treatment, wound healing, cholesterol lowering or antibody dependent cellular cytotoxicity (ADCC) in mammals comprising administering to the mammal an effective amount of a pharmaceutical preparation comprising the conjugated CSF-1 protein dissolved in a pharmaceutically acceptable aqueous carrier medium.
  • ADCC antibody dependent cellular cytotoxicity
  • Figure 1 shows the cDNA and deduced amino acid sequence for pCSF-17 (32 amino acid leader sequence and amino acids 1-224).
  • Figure 2 shows the cDNA and deduced amino acid sequence for CSF-4 (partial amino acid leader sequence and amino acids 1-522).
  • Figure 3A shows the size exclusion HPLC chromatogram of underivatized recombinant CSF-1 (rCSF-1) (SCSF/NV2CV150).
  • Figure 3B shows the HPLC chromatogram of the same rCSF-1 derivatized with PEG- NHS of 7000 dalton average molecular weight.
  • Figure 4 shows a size exclusion HPLC chromatogram of rCSF- 1 (SCSF/CV150) derivatized with PEG-NHS of 11,000 daltons average molecular weight.
  • Figure 5 shows a graph of CSF-1 concentration in rat blood plasma versus time for R g ⁇ ji rCSF- 1 (SCSF/CV150), the dimeric product derived from the rCSF-1 with the sequence in Figure 2 [lacking the leader sequence and a C-terminal sequence, but retaining essentially all of the N- terminal methionine present in the E. c ⁇ ji construct], rCSF-1 expressed in mammalian cells (COS) that arises from a glycosylated 522-amino acid precursor (LCSF) andPEG-ll.OOO-derivatized R coH rCSF-l (SCSF/CV150).
  • COS mammalian cells
  • Figure 6 shows a size exclusion HPLC chromatogram of rat blood plasma 120 minutes after intravenous injection of PEG- 11,000- de ⁇ ivatizedrCSF-1 (SCSF/CV150). Radioimmunoassay (RIA) and absorbance at 280 nm are plotted.
  • RIA Radioimmunoassay
  • Figure 7 shows a graph of CSF-1 concentration in rat blood plasma versus time for K coti rCSF-1 (SCSF/NV3C 158) and for the same protein derivatized with 11,000 PEG.
  • Figure 8 shows SDS-PAGE analysis of rCSF-1 derivatized with PEG-11,000 (SCSF/CV150). Gel (10%), stained for protein with
  • Coomassie blue is of the sample used in Figure 5, with and without reduction of disulfide bonds.
  • Colony stimulating factor- 1 refers to a dimeric protein or glycoprotein composition that has the biological activity for CSF-1 in the standard in vitro colony stimulating assay of Metcalf. J. Cell. Physiol. (1970) 26:89 wherein the polypeptide stimulates the formation of primarily macrophage colonies.
  • Biologically active CSF-1 means a composition of conjugated CSF-1, that has essentially the same specific activity in mouse bone marrow colony-forming assays as the unconjugated form of the same CSF-1 or at least about 10% of its specific activity.
  • a "native" CSF-1 is a non- recombinant CSF-1.
  • Murine CSF-1 is described in European Patent Publication No. 0272779, Rajavashisth et al., Proc. Natl. Acad. Sci. USA (1987) Mil 157 and in DeLamarter et al., Nucleic Acids Res. (1987) 11:2389.
  • CSF-1 "Clinically pure” CSF-1 means a preparation of biologically active human CSF-1 produced recombinantly in bacteria that is at least 95% CSF-1 either by RP-HPLC or by either reducing or non-reducing SDS-PAGE analysis and has an endotoxin content of less than about 1.0 ng/mg CSF-1.
  • “Selectively conjugated” refers to proteins that are covalently bonded via free amino groups of the protein (preferably one or two free amino groups, to retain maximum biological activity), via free sulfhydryl groups (if any), or via a carbohydrate moiety (if any) present in the protein.
  • “Effective amount” and “immunotherapeutically effective amount” signify an amount effective to perform the function specified, such as to promote tumor reduction or prevent or cure infectious diseases. The exact optimal amount will depend on many factors, including the patient's clinical history and current disease, die schedule, the route, and the response of the patient
  • “Therapeutic treatment” indicates treating after the disease is contracted, whereas “prophylactic” treatment indicates treating to prevent contraction of the disease.
  • “Mammals” indicates any mammalian species, and includes rabbits, mice, dogs, cats, cattle, sheep, primates, and humans, preferably humans.
  • Meats are genetically engineered proteins expressed from a nucleic acid sequence that has been altered using techniques such as site-specific mutagenesis. Such genetic alterations are designed to result in one or more substitutions, additions, and/or deletions to the amino acid sequence of the parent protein.
  • “Pharmaceutically acceptable” refers to a carrier medium that does not interfere with the effectiveness of the biological activity of the active ingredient(s), is stable, and is not toxic to the host to whom it is administered.
  • “Homodimer” refers to a dimeric protein essentially identical in its two subunits except that it also includes dimeric proteins with minor microheterogeneities that occasionally arise on production or processing of recombinant proteins.
  • ' ⁇ eterodimer refers to a dimeric protein having subunits which differ in amino acid sequence, the number of amino acids or both. In other words, heterodimers have two nonidentical subunits.
  • CSF-1 Proteins .
  • CSF-1 is biologically active in its dimeric form.
  • the CSF-1 employed herein may be the native dimer or recombinantly produced dimer.
  • Native dimeric CSF-1 species have been obtained from human urine, cultured monocytes, and culture supematants of some cell lines. It has been possible to obtain recombinant DNA encoding CSF-1 monomers consisting of a variety of amino acid sequences and lengths.
  • Figures 1 and 2 show the DNA sequences and amino acid sequences predicted from the DNA sequences for, respectively, the full- length, unprocessed short and long forms, both of which contain a 32-amino acid signal sequence at their N-termini.
  • sequences of the monomeric CSF- 1 protein are considered herein for convenience to be the 224-amino-acid short form without the leader sequence (designated herein as SCSF), and the 522- amino-acid long form without the leader sequence (designated herein as LCSF).
  • SCSF 224-amino-acid short form without the leader sequence
  • LCSF 522- amino-acid long form without the leader sequence
  • Other CSF-1 dimers produced from DNA sequences modified by point mutation(s), insertion(s), deletion(s), and/or translocation(s) are also expected to benefit from the chemical modification of this invention if they are biologically active.
  • the recombinant CSF-1 produced in any host, whether eukaryotic or prokaryotic, may be conjugated to polymers via selected amino acid side groups, preferably free amino groups.
  • the DNA encoding CSF-1 is expressed in bacteria and the resulting CSF-1 is a homodimer after purification and refolding. If the conjugation is via a carbohydrate moiety, the host may be eukaryotic, or glycosylation may be carried out in vitro.
  • LCSF denotes the 522-amino acid sequence disclosed for the clone CSF-4, set forth in European Patent Publication No. 0272779 referred to above and shown in Figure 2, without the complete signal sequence.
  • SCSF denotes the 224-amino acid sequence disclosed for the clone pCSF-17, shown in Figure 1, without the signal sequence, described in the Kawasaki article referred to above, Science (1985) 220:292-296, also incorporated herein by reference. It will be noted that one particular pCSF-17 clone derivative has an aspartic acid residue at position 59.
  • the disclosed LCSF clone also encodes Asp at position 59. Muteins corresponding to amino acid substitutions within the sequences depicted in Figures 1 and 2 are correspondingly designated by the substitution subscripted with the position. Mutein forms of CSF-1 are disclosed in European Patent Publication No. 0272779 published June 29, 1988, the disclosure of which is incorporated herein by reference. When constructs encoding these proteins are expressed in bacteria, the final products may also retain N-terminal methionine to varying degrees. Since the extent of N-terminal methionine removal cannot be reliably predicted, this possibility is not included in the notation.
  • LCSF sequences will be designated as CV or NV, respectively.
  • the C- terminal deletions will be followed by the number representing the number of amino acids of the native structure remaining; for the -N-terminal deletions,
  • NV will be followed by the number of amino acids deleted from the N- terminus.
  • LCSF/C 150 denotes a construct encoding a protein that contains the first 150 amino acid residues of the long CSF sequence
  • SCSF/CV158 denotes a construct encoding a protein that contains the first 158 amino acid residues of the short form
  • SCSF/NV2 denotes a construct encoding the short form with two N-terminal amino acids deleted.
  • LCSF/NV2CV150 denotes a form that is the same as LCSF/CV150, except that the two N- terminal glutamic acid residues are deleted.
  • Plasmids encoding a variety of CSF-1 forms are currently available, and can be expressed in bacterial systems.
  • a form of plasmid encoding the long form of CSF-1 can be expressed in eukaryotic cells, in which case, the eukaryotic cell "processes" the clone, secreting a protein that is C-terminally truncated and has the leader sequence removed from the N- terminus.
  • the clone can be truncated to express specific C- terminally deleted forms in eukaryotic or prokaryotic cells.
  • the first two or three N-terminal codons can be deleted so that the resulting protein expressed in E. coli is more homogeneous.
  • N-terminal methionine encoded just upstream of the mature native sequence N-terminus in the E. coli constructs (which is retained in the protein as "N-terminal Met" unless removed by post-translational processing) is more readily removed from these N-terminal deletion constructs.
  • significant heterogeneity detectable using reverse-phase HPLC (RP-HPLC) is found when the gene encoding the "native" N-terminal sequence (for example, of the short form, mutein SCSF/CV150) is expressed in E. coli and the purified product is characterized.
  • This heterogeneity is ehminated when the corresponding CSF-1 gene lacking the two N-terminal codons (glutamic acid) is expressed.
  • N-terminal truncated forms of other short and long CSF-1 gene constructs can also be employed.
  • Preferred constructions are those wherein the protein consists essentially of an amino acid sequence selected from the group consisting of
  • LCSF/CV150 through CV464, tyr 59 LCSF/C 150 through CV464; asp 5 -SCSF/CV150 through CV166. More preferred are those CSF-1 proteins consisting essentially of an amino acid sequence selected from the group consisting of LCSF/CV150; LCSF/CV190; LCSF/CV221;
  • LCSF/CV258 LCSF/CV406; LCSF/CV411; LCSF/CV464; asp 59 SCSF/CV150; asp 5.
  • LCSF/CV221, asp 59 SCSF/CV158, asp 59 SCSF/CV150 and their corresponding NV2 and NV3 forms are preferred starting materials.
  • the most preferred starting materials are the products of the clones encoding asp 59 SCSF/CV150; asp 59 SCSF/CVl58; LCSF/CV221 and their corresponding NV3 forms.
  • the CSF-1 may be in the form of a heterodimer prepared from among various monomeric units of CSF-1, particularly if the conjugation is done through a reactive cysteine residue of the CSF-1 heterodimer.
  • the large number of CSF-1 proteins formed by variations in C- terminal sequence due either to differences in processing or clone construction provides a variety of starting materials that can be utilized in heterodimer formation.
  • novel heterodimeric materials can be formed by refolding.
  • the monomeric form of SCSF/CV150 along with the monomeric form of LCSF/CV157, can be mixed and treated according to the method of PCT Publiction No. WO 88/08003 published October 20, 1988, the disclosure of which is incorporated herein by reference.
  • the heterodimer can then be separated from the homodimeric and oligomeric side products by various chromatographic and other methods.
  • the heterodimer (particularly
  • SCSF/CV150 and LCSF/CV157 can be made to terminate on one subunit at the 157 position, providing a potentially free sulfhydryl group for reaction with the conjugation polymer.
  • Similar mixtures subjected to the method of the invention could lead to heterodimers of components having amino acid substitutions ⁇ e.g., glu 52 LCSF/CV221 andLCSF/C 190.
  • the differing monomers may be mixed in vitro or produced in the same cell. If produced in the same cell, a construct for expression of each monomer is introduced into the same host; in such embodiments, it is preferred that each construct bear a different marker (such as tetracycline resistance (Tc R ) and Ampicillin resistance (AmpR), so that cotransformed hosts are selected. The cotransformed cells are then grown and induced to obtain mixtures of the two forms.
  • Tc R tetracycline resistance
  • AmpR Ampicillin resistance
  • single cysteine residues can be engineered into CSF-1 in a non-natural location to create a construct of rCSF-1 containing a free sulfhydryl group on a cysteine residue for PEG reaction, provided the molecule retains bioactivity.
  • Heterodimer constructs containing a substitution of a given cysteine in only one of the two subunits may also be useful in the generation of free sulfhydryls.
  • carbohydrate moieties can be placed on the CSF-1 protein, either by expression from eukaryotic systems or by in vitro glycosylation with enzymes.
  • CSF-1 proteins may or may not retain the exact length prescribed by the clone, due to processing by the host Therefore, although the starting material proteins are referred to by the same designation, it should be understood that these designations, in reality, refer to the clone construct and the actual length of the starting material for the process disclosed herein may be shorter, or longer (if it has N-terminal Met), than that specified by the C-terminal amino acid number.
  • the CSF-1 may be produced in eukaryotic cells, e.g., mammalian, insect, or yeast cells. Production in mammalian cells is described in PCT Patent Publication No. WO 86/04607.
  • the CSF-1 may be produced from insect cells using a baculovirus vector, as described in Luckow et al., Biotechnol. (1988) 6:47, the disclosure of which is incorporated herein by reference. Other descriptions of this type of CSF-1 production are contained within Weaver et al., BioTechnologv (1988) 6:287.
  • Insect-cell-produced CSF-1 (SCSF/CV150) is a glycosylated dimer that was found to have a very short in vivo half-life.
  • the two N-linked glycosylation signals in the amino acid sequence of SCSF (Asn 122 and Asn 140) could be modified in the DNA using site-directed mutagenesis to generate a non- glycosylated, bioactive insect-cell-produced CSF-1 protein (Gin 122, Gin 140). This protein can then be reacted via its lysine or cysteine residues with a polymer as done with prokaryote-expressed rCSF-1 to increase its in vivo half- life.
  • the polymer can be attached covalently to the glycosylated CSF-1 on the carbohydrate, by reacting, for example, PEG-amine, PEG- hydrazine, or PEG-hydrazide with CSF-1 protein that has been oxidized with periodate to convert vicinal diols of the sugars to aldehydes.
  • the PEG-amine can be prepared by converting PEG-OH to PEG-tosylate and then to PEG- phthalimide, and the phthalimide is cleaved with hydrazine to produce PEG- NH 2 in a Gabriel synthesis.
  • PEG-hydrazine and PEG-hydrazide can be prepared by methods known to those skilled in the art.
  • the PEG derivative will couple at carbohydrate sites where vicinal diols were converted to aldehydes.
  • CSF-1 is produced intracellularly in bacterial hosts, it must be refolded to form active dimers in high yield, as described in PCT Publication No. WO 88/08003 supra.
  • the solubilized monomer is maintained in a chaotropic environment under reducing conditions.
  • a suitable reducing agent such as ⁇ - mercaptoethanol or dithiothreitol (DTT).
  • DTT dithiothreitol
  • the solubilized protein is typically maintained in, for example, 8 M urea or 7 M guanidinium hydrochloride, at a pH of about 7-8.6, in the presence of about 2-100 mM thiol compound.
  • the monomer may either be refolded directly or purified from remaining proteins by a suitable purification procedure such as chromatography on an adsorbant gel, chromatography using an ion exchange column, or gel-permeation chromatography prior to refolding.
  • Gel-permeation chromatography is useful, as it permits an easy size separation of the desired monomer length, which is generally known in advance, from impurities of differing molecular weights.
  • DEAE Sepharose chromatography in urea is even more useful, as it concentrates as well as purifies and is more easily scaled up than is sizing. It is preferable that the purification be conducted under reducing conditions to prevent the formation of disulfide-linked aggregates.
  • a suitable reducing agent is included in the solutions used to load the chromatographic columns or batches and in the eluting solutions.
  • low pH may be substituted for reducing agent, as low pH will essentially prevent disulfide bond formation and is compatible with some chromatographic systems.
  • inclusion of a low concentration of a chelating agent may help to preserve the reduced form of the protein.
  • the partially purified monomer is then subjected to refolding conditions for the formation of the dimer.
  • the protein concentration during this step is of considerable importance. Generally, final percent yields per volume of the CSF-1 refolding reaction are increased if the protein concentration is less than about 2 mg ml of the CSF-1 protein; a concentration range of 0.03-1.0 mg/ml is preferred.
  • the refolding conditions may include gradual removal of the chaotropic environment over an appropriate time period, usually several hours, or dilution of the sample to the desired concentration of protein and chaotropic agent. Also possible are methods that provide a constant protein concentration, such as dialysis or hollow fiber diafiltration, while the chaotrope is slowly removed.
  • a relatively nondenaturing level is reached.
  • guanidine hydrochloride is used as a chaotrope, a final concentration of less than about 2 M, and preferably 0.1-1 M, is attained, and if urea is used as the chaotrope, a final concentration of less than about 1 M, and preferably 0.1-0.5 M, is attained.
  • the refolding is conducted so as to permit oxidation of the sulfhydryl groups to disulfides that establish the biologically active dimeric configuration that, in the case of CSF-1, requires the formation of disulfide bonds, one or more of which links the two chains.
  • One or more intrachain disulfides are also formed.
  • Suitable redox conditions that encourage this formation of dimers include the sulfhydryl/disulfide reagent combinations, such as oxidized and reduced glutathione.
  • the ratio of reduced to oxidized glutathione or other sulfhydryl/disulfide combination is typically from about 2 mM/0.1 mM to 0.5 mM/1.0 mM. Other methods of oxidation are also acceptable.
  • the pH of the solution during the refolding process should be maintained at about 7.5 to 9.0 to favor the reaction. It is clear that during the process of refolding die highly reducing conditions under which the initial purification was conducted are no longer employed. The exclusion of significant concentrations of salts, such as sodium chloride, during the refolding process, permits the use of ion exchange chromatography as a subsequent concentration/purification step.
  • Residual redox reagents present in die refolded CSF-1 preparation may possibly facilitate disulfide exchanges during subsequent purification steps.
  • Two more preferred procedures to block such unwanted disulfide exchanges include lowering the pH to below 7.0 or diafiltration to remove the redox reagents.
  • removal of the redox material and concentration of the refolded proteins may be performed by direct loading of the refolded material onto an ion exchange chromatography column using, for example, DEAE Sepharose. Frequendy such procedures are carried out at pHs around 8; however, lowering the pH into the range of 5.5 to 7.0 reduced oligomer formation and increased yield of dimeric CSF-1.
  • the dimer is further purified from residual redox material and from other proteins using procedures similar to those set forth above for the monomer.
  • Suitable means include gel filtration, hydrophobic interaction chromatography, ion exchange chromatography, and reverse-phase HPLC.
  • a particularly successful protocol for removal of undesirable impurities such as pyrogens or other endotoxins includes the use of chromatography on a phenyl-TSK or phenyl-Sepharose column.
  • the chromatography is carried out under conditions and with reagents that are essentially endotoxin-free.
  • the desired dimeric CSF-1 is soluble and stable in approximately 1.5 M ammonium sulfate at neutral pH, and is loaded onto d e columns under these conditions at low temperatures, i.e., from about 4°C to about 10°C, and preferably about 4°C. Removing the precipitate that forms on adding die ammonium sulfate removes some aggregates and unstable forms of refolded CSF-1.
  • the CSF-1 protein can be eluted using a gradient of decreasing ammonium sulfate (1.5 to 0 M) with increasing ethylene glycol (0 to 50%) in neutral buffer.
  • the CSF-1 dimer elutes at approximately 0.6 M ammonium sulfate, 35% ethylene glycol from the phenyl-TSK column.
  • Propylene glycol may be used instead of ethylene glycol, in which case the elution conditions will be somewhat different Alternative supports can also be used, andphenyl-Sepharose is, in fact preferred for larger scale production of die purified CSF-1 dimeric protein.
  • the CSF-1 protein described above is conjugated to die polymer via either (1) free amino group(s), preferably only one or two in order to minimize loss of biological activity, (2) at least one carbohydrate moiety on the protein, or (3) free sulfhydryl group(s) that is/are engineered into d e clone and remain free after refolding.
  • the number of polymer molecules tiiat have been conjugated to the protein can be determined by various methods, including, for example, acid degradation or digestion, followed by amino acid analysis if the links are maleimido or bromoacetyl to cysteine links and die extent of derivatization is high (more than 4 moles/mole).
  • the conjugated protein can be digested into small fragments with an enzyme (e.g., trypsin) and separated by column chromatography. A peptide map of d e protein before and after modification would be compared, and fragments with altered elution times sequenced to determine the location(s) of polymer attachments.
  • the polymer can be radioactively labeled prior to coupling to determine how many moles of radioactive polymer are attached per mole of CSF-1 protein.
  • polymers of relatively uniform molecular weight are conjugated to CSF-1 of uniform molecular weight measurement of die molecular weight of the conjugate can serve as an estimate of me number of polymers per CSF-1 molecule.
  • the residue(s) to be conjugated may be: (1) any free amino groups ( ⁇ -amino groups at lysine residues or a free amine group, if any, at the
  • the polymer to which the protein is attached is a homopolymer of polyethylene glycol (PEG) or of polypropylene glycol (PPG), a polyoxyethylated polyol, or polyvinyl alcohol, provided in all cases that the polymer is soluble in water at room temperature.
  • PEG polyethylene glycol
  • PPG polypropylene glycol
  • polyoxyethylated polyol or polyvinyl alcohol, provided in all cases that the polymer is soluble in water at room temperature.
  • polyoxyediylated polyols include polyoxyethylated glycerol, polyoxyethylated sorbitol, polyoxyediylated glucose, and die like.
  • the glycerol backbone of polyoxyethylated glycerol is die same backbone occurring naturally in, for example, animals and humans in mono-, di-, and triglycerides. Therefore, this compound might not necessarily be seen as foreign in the body.
  • the polymer preferably has an average molecular weight between about 1000 and 100,000 daltons, more preferably between 4000 and 40,000 daltons, depending, for example, on the molecular weight of the particular CSF-1 employed. Since the object of the modification is to obtain a conjugated protein witii retained biological activity, witii enhanced in vivo half- life over the unconjugated protein, and witii reduced immunogenicity, the molecular weight of the polymer will be chosen to optimize these conditions, e.g., a modified dimeric CSF-1 protein of over about 80 Kd apparent molecular weight. An additional advantage gained in derivatizing native dimeric
  • CSF-1 i.e., non-recombinant protein
  • CSF-1 is tiiat the use of a scarce CSF-1 that is hard to purify would be maximized by such a modification.
  • the PEG homopolymer is substituted at one end witii an alkyl group, but it may also be unsubstituted.
  • the alkyl group is a -C 4 alkyl group, and most preferably a methyl group.
  • the polymer is a monomethyl-substitutedPEG homopolymer and has a molecular weight of about 4000 to 40,000 daltons.
  • the covalent modification reaction may take place by any of the methods described above, preferably at about pH 5-9, more preferably 7-9 if 5 the reactive group on the protein is a free amino group.
  • the protein is conjugated via at least one terminal reactive group added to die polymer.
  • the polymer with die reactive group(s) is designated herein as "activated polymer”.
  • the reactive group(s) selectively react with a free amino or other reactive group of the protein. (If there is more than one
  • the amount of intact activated polymer employed is generally about 1- to 30-fold molar excess of the active polymer over the protein. The exact molar ratio used is dependent upon a number of variables including die type of activated
  • d e molar excess of activated polymer over protein is preferably no more than about 11 moles per mole of CSF-1 dimer for derivatization of amino groups, and most preferably is about l to 8 moles per mole of CSF-1.
  • the molar ratio of activated polymer over protein is preferably no more than about 11 moles per mole of CSF-1 dimer for derivatization of amino groups, and most preferably is about l to 8 moles per mole of CSF-1.
  • PEG-chloroformate active ester PEG-PNP
  • excess of PEG-chloroformate active ester (PEG-PNP) over protein is preferably no more than about 5 moles per mole of CSF-1 dimer for derivatization of amino groups and most preferably is about 1 to 2 moles per mole of CSF-1.
  • reaction is carried out in a buffer of pH about 7-9, frequendy at about 10 mM Hepes pH 7.5, 100 mM NaCl, if an internal-ester-free PEG-NHS reagent is used or at about 50 mM Na borate pH 9 if the PEG-PNP reagent is used, bodi of which are described below.
  • the reaction is carried out generally at 0 to
  • the modification reaction with active PEG can be performed in many ways, described below, using one or more steps. Examples of suitable
  • 35 modifying agents that can be used to produce the activated PEG in a one-step reaction include cyanuric acid chloride (2,4,6-trichl ⁇ ro-S-triazine) and cyanuric acid fluoride.
  • die modification reaction takes place in two steps wherein die PEG-OH is reacted first with an acid anhydride such as succinic or glutaric anhydride to form a carboxylic acid, and die carboxylic acid is tiien reacted with a compound capable of reacting with the carboxylic acid to form an activated PEG witii a reactive ester group that is capable of reacting witii the protein.
  • acid anhydride such as succinic or glutaric anhydride
  • die carboxylic acid is tiien reacted with a compound capable of reacting with the carboxylic acid to form an activated PEG witii a reactive ester group that is capable of reacting witii the protein.
  • an acid anhydride such as succinic or glutaric anhydride
  • die carboxylic acid is tiien reacted with a compound capable of reacting with the carboxylic acid to form an activated PEG witii a reactive ester group that is capable of
  • monomethyl-substituted PEG may be reacted at elevated temperatures, preferably about 100-110°C for four hours, with glutaric anhydride.
  • the monomethyl PEG-glutaric acid ti s produced is tiien reacted with N-hydroxysuccinimide in the presence of a carbodiimide reagent such as dicyclohexyl or diisopropyl carbodiimide to produce d e activated polymer, metiioxypolyethylene glycolyl-N-succinimidyl glutarate, which can tiien be reacted with the protein after purification.
  • a carbodiimide reagent such as dicyclohexyl or diisopropyl carbodiimide
  • the monomethyl-substituted PEG may be reacted with glutaric anhydride followed by reaction with 4-hydroxy-3- nitrobenzene sulfonic acid (HNS A) in the presence of dicyclohexyl carbodiimide to produce d e activated polymer.
  • HNSA 4-hydroxy-3- nitrobenzene sulfonic acid
  • die PEG may be activated for attachment to the protein using PEG-amine or PEG-OH as starting materials.
  • the PEG- OH may be converted to die PEG-amine as described by V.N.R. Pillar et al., J. Organic Chem.. 45_:5364-5370 (1980), the disclosure of which is incorporated herein by reference.
  • monomethyl PEG-amine (mPEG) is prepared by converting mPEG-OH to mPEG-tosylate and then to mPEG- phthalimide, and the phthalimide is cleaved with hydrazine to produce mPEG- NH 2 in a Gabriel synthesis.
  • the mPEG-amine is then reacted witii glutaric anhydride at room temperature for about four hours to produce mPEG- NHCO((2_ 2 ) 3 COOH. After the reaction the product is precipitated, purified, and reacted with N-hydroxysuccinimide and
  • This compound can then be reacted witii the appropriate free amino group(s) of the CSF-1 polypeptide.
  • active ester forms of polyethylene glycol carboxylic acid useful for such conjugation are described in NitecM et al., Peptide Chemistry 1987 , Shiba & Sakakibara (Ed). Protein Research Foundation, Osaka (1988). Briefly, the active esters have a formula of:
  • esters may be produced, for Compound I, by alkylation of the polyethylene glycol with an alpha-haloalkanoic acid or ester thereof followed by esterification with HO-CH 2 -CN or the group corresponding to LG, or, for Compound (II), by oxidation of d e PEG to its acid, followed by esterification with HO-CH 2 -CN or the group corresponding to LG.
  • the activating agent is para-nitrophenol or ortho- nitrophenol.
  • the polymer is conjugated to die protein via an amide linkage formed from die para-nitrophenyl ester of the polymer.
  • the PEG-OH may be converted to PEG-O- and reacted with BrCH 2 C0 2 CH 3 , the methyl ester may be hydrolyzed, and die acid may be reacted witii j2-nitrophenol in the presence of dicyclohexyl-carbodiimide to produce PEG-0-CH 2 COO- ⁇ NO 2 .
  • This polymer is, following purification, in turn reacted with available free amino group(s) of the CSF-1.
  • PEG-OH is reacted witii a chloroformate (also called a chlorocarbonate) to form a PEG active ester also called (PEG-PNP).
  • the chloroformate is made by reacting phosgene also known as carbonyl chloride with an alcohol (R-OH) which contains electron withdrawing substituents on the carbon that carries the -OH.
  • the alcohol is preferably an acidic alcohol, more preferably an acidic alcohol which contains aromatic rings which have high extinction coefficients.
  • R groups are: N-hydroxy-succinimide, N-hydroxy-sulfosuccinimides, cyanomethyl esters, all nitro, chloro, and cyano substitutions on benzene, naphthalene, or larger aromatic ring systems which may or may not contain hetero-atoms, such as pyridine, para-nitrophenol (PNP), ortho-nitrophenol (ONP), etc.
  • Most preferred R groups are PNP and ONP.
  • the chloroformate is reactive at two sites; at the bond between the chlorine and carbonyl group (more reactive) and die bond between the carbonyl and O-R group (less reactive).
  • the more reactive site is where the chloroformate binds to the PEG.
  • PEG-OH and the chloroformate are preferably added together at room temperature in an appropriate solvent, such as CHCI 3 , or CH 2 CI 2 .
  • an acylation catalyst is added between 0 and 1 hours later, preferably the catalyst is pyridine or dimethyl pyridine.
  • the chloroformate is added up to 12 M excess, more preferably to a 2 M excess. The mixture is allowed to mix for preferably 4 hours, more preferably 16 hours. At this point, a precipitate may form.
  • the resulting solution contains the PEG active ester as well as unreacted PEG and excess chloroformate. It is precipitated by adding an etiier, preferably the ether is dietiiyl ether. The precipitate contains the PEG active ester and can be washed witii appropriate solvents such as ether, redissolved and reprecipitated if necessary.
  • the chloroformate portion of the PEG active ester still has the less reactive site available. At this site, the covalent bond between the PEG and CSF-1 is formed. In the final product the PEG moiety is bound to CSF-1 by a
  • urethane also called a carbamate, linkage ha ing the structure -0-C-NH-.
  • This linkage is relatively stable and will keep PEG conjugated to CSF-1 with little or no hydrolysis under physiological conditions.
  • Homodimeric recombinant CSF-1 from E. coli does not contain significant numbers of reactive free sulfhydryl groups after refolding has correctly proceeded to completion.
  • the clone could be modified genetically, however, to include one or more novel cysteine residues that might retain sulfhydryl groups following refolding.
  • the CSF-1 mutein so produced must still retain significant biological activity to be useful herein.
  • free sulfhydryls can be generated by creation of selected heterodimers or by partial refolding of homodimers such that certain
  • SH groups such as on cysi 59 , ⁇ e available for modification by activated polymers.
  • a preferred mode of conjugation is as follows: mPEG-NH 2 as described above is reacted at room temperature for preferably 0.5-1.5 hours with N-maleimido-
  • mal-sac- HNSA 6-aminocaproic ester of 4-hydroxy-3-nitrobenzene sulfonic acid
  • the latter reaction is preferably conducted with about a 5-fold molar excess of mal-sac HNSA over PEG-NH 2 .
  • the reagent can then be reacted with the protein at room temperature in a buffer using equimolar amounts of reagent and protein.
  • reagents such as N-succmimidyl-4-(N-maleimidomed ⁇ yl)-cyclohexane-l- carboxylate (SMCC), or X ⁇ _ 2 CO-NH(CH 2 ) 5 -HNS A ester wherein X is Br, Cl, or I, can perform the same function as mal-sac HNSA under a variety of reaction conditions known to those skilled in die art.
  • SMCC N-succmimidyl-4-(N-maleimidomed ⁇ yl)-cyclohexane-l- carboxylate
  • X ⁇ _ 2 CO-NH(CH 2 ) 5 -HNS A ester wherein X is Br, Cl, or I can perform the same function as mal-sac HNSA under a variety of reaction conditions known to those skilled in die art.
  • Commonly used salts are ammonium sulfate, (NH 4 ) 2 S0 4 ; sodium sulfate, Na 2 S ⁇ 4 ; magnesium salts and phosphates.
  • NH 4 ) 2 S0 4 ammonium sulfate
  • sodium sulfate sodium 2 S ⁇ 4
  • magnesium salts and phosphates The more highly conjugated species precipitated at lower salt concentrations than did less conjugated and unconjugated protein. Recycling ofpartially purified species through any of these techniques may result in a substantially homogeneous species of conjugated protein with regard to die number of polymers per molecule of protein.
  • compositions The protein thus modified and optionally purified as to extent of conjugation is then formulated in a non-toxic, stable, pharmaceutically acceptable aqueous carrier medium, preferably at a pH of about 3 to 8, more preferably 5-8.
  • Administration is by conventional protocols and regimens, preferably systemic, including intravenous administration.
  • Aqueous formulations compatible with the culture or perfusion medium will generally be used.
  • the composition may include conventional physiologically acceptable excipients, such as water for injection, buffers, and stabilizers, as are known in the art
  • a water-soluble carrier such as mannitol may optionally be added to die medium.
  • Modified CSF-1 may be used eitiier as the sole active ingredient or in combination with other proteins or compounds having complementary activity.
  • Such other compounds may include antitumor agents such as adriamycin of lymphokines such as D -1, -2, -3, -4, interferons (alpha, beta or gamma), GM-CSF; G-CSF, and tumor necrosis factor.
  • antitumor agents such as adriamycin of lymphokines such as D -1, -2, -3, -4, interferons (alpha, beta or gamma), GM-CSF; G-CSF, and tumor necrosis factor.
  • the effect of the modified CSF-1 may be improved by the presence of such additional components.
  • a summary of formulation techniques for pharmaceutical compositions, including protein, is found, for example, in Remington's Pharmaceutical Sciences. Mack Publishing Co., Easton, PA, latest edition.
  • the dosage level of protein in the formulation will depend on die in vivo efficacy data obtained after preclinical testing and may vary depending upon die clinical application.
  • the medium in which the protein is dissolved will be at a pharmaceutically acceptable pH when the mixture is reconstituted.
  • the lyophilized mixture may be reconstituted by injecting into the vial a conventional parenteral aqueous solvent such as, e.g., distilled water for injection.
  • a conventional parenteral aqueous solvent such as, e.g., distilled water for injection.
  • the reconstituted formulation prepared as described above is suitable for parenteral administration to humans or other mammals in therapeutically effective amounts (i.e., amounts that eliminate or reduce the patient's pathological condition witiiout mortality or unacceptable morbidity) to provide tiierapy thereto.
  • CSF-1 therapy may be appropriate for a variety of indications such as enhancing the immune system, enhancing cytotoxicity of macrophages, increasing monocytic and granulocytic white blood cell count treating infectious diseases such as cytomegalovieris and bacterial infections (e.g., Gram-negative sepsis) by therapeutic or prophylactic administration to mammals, treating tumor burden such as sarcoma or melanoma in mammals, treating cholesterolemia (as described for GM-CSF by Nimer et al (1988) JAMA 260:3297-3300) and/or healing wounds in mammals.
  • CSF-1 may be combined with G-CSF for stimulation of the immune system, as described in European Patent Publication No.
  • the dose and dosage regimen of the conjugated CSF- 1 will depend, for example, upon the pha ⁇ nacokinetics of the drug, the nature of the disease or condition, the type and length of polymer, the characteristics of the particular CSF-1, e.g., its therapeutic index, its spectrum of activities, the patient, and die patient's medical history. Different modified CSF-1 proteins are expected to have different pharmacokinetic and tiierapeutic properties that are advantageous for different routes of administration. A long-acting drug might only be administered every 3-4 days, every week, or once every two weeks. The clearance rate can be varied to give ultimate flexibility to fit the particular need of die patient by changing, e.g., the type of polymer, the size of the polymer attached, and the amino acid sequence to which the polymer is attached.
  • a linear ester of monomethyl PEG of average molecular weight 7000 can be obtained by first reacting monomethyl PEG-7000, which is available from Union Carbide, widi glutaric anhydride at 100 to 110°C for four hours or by a method similar to that of Abuchowski et al.. Cancer Biochem. Biophvs..2:175-186 (1984), the disclosure of which is incorporated herein by reference.
  • the resulting PEG-glutarate was reacted with N-hydroxysuccinimide in the presence of dicyclohexylcarbcdiimide, as described in detail by Abuchowski et al.. supra, on page 176.
  • PEG*-7000 metiioxypolyethylene glycolyl N-succinimidyl glutarate, hereinafter designated as PEG*-7000.
  • a PEG-11,000 glutaramido NHS species (PEG"- 11,000) was prepared as follows, according to die procedure of Rajasekharam Pillai et al., J. Ore. Chem.
  • the mPEG-tosylate (about 1.5 mmole) was dissolved in 20 ml dimethylformamide, and 2.5 g (17.0 mmole) potassium phthalimide was added. The solution was heated at reflux under nitrogen for four hours.
  • the mPEG-tosylate (about 1.5 mmole) was dissolved in 20 ml dimethylformamide, and 2.5 g (17.0 mmole) potassium phthalimide was added. The solution was heated at reflux under nitrogen for four hours. The
  • the precipitate was collected by filtration and resuspended in
  • the cells were harvested by 5-fold concentration and diafiltered against ten volumes of 5 mM EDTA, pH 8.5, using Dorr-Oliver tangential cross-flow microporous filtration.
  • the cells were disrupted by ti ree passes at 7,500 psi in a Manton-Gaulin high pressure mechanical cell homogenizer. 1- Octanol was added to 0.1% (v/v) and d e homogenate was held overnight at 4°C.
  • the homogenate was made 25% sucrose by addition of a 63% w/v sucrose solution.
  • the insoluble protein fraction (refractile bodies) was separated from cell debris by continuous flow disk stack centrifugation (Westphalia SB7) at 9000 xg, 1 liter/minute and 4-6°C.
  • the wet pellet was mixed 50:50 (w/v) with deionized water and stored at -20°C in aliquots. Twenty-five grams of refractile body suspension
  • a 150-mI aliquot of the solubilized CSF-1 was tiien loaded onto a 5 x 8 cm DEAE-Sepharose (Pharmacia) column equilibrated in 6 M urea containing 25 mM Tris, 10 mM sodium phosphate buffer (pH 7.0). The column was washed with 1 bed volume of die above solution, which had been modified to contain 1 mM DTT and 1 mM EDTA, and the CSF-1 was then eluted with a 1.4-liter salt gradient of 0-0.6 M sodium chloride in the wash buffer. The CSF-1 peak eluted at approximately 0.06 M sodium chloride.
  • the remaining 90 ml of solubilized refractile bodies was tiien purified over die DEAE-Sepharose column in identical fashion.
  • the combined CSF-1 pools (165 ml) contained approximately 250 mg of protein at a purity of approximately 50%.
  • the CSF-1 was then refolded by diluting die DEAE-pool into refolding buffer containing 50 mM Tris (pH 8.5), 5 mM EDTA, 2 mM reduced glutathione, 1 mM oxidized glutathione, pre-cooled to 4°C.
  • the CSF- 1 was allowed to refold for 30 hours at 4°C.
  • the pH of the refolded CSF-1 was tiien adjusted to 6.8 using 8.5% phosphoric acid solution.
  • the solution was then clarified by centrifugation for 10 minutes at 15,000 x g and loaded onto a 5 x 4 cm DEAE Sepharose column pre-equilibrated in 10 mM sodium phosphate, 25 mM Tris (pH 6.8).
  • the column was washed with 300 ml of this buffer and tiien eluted witii a 700 ml, 0-0.6 M sodium chloride gradient in die same buffer system.
  • the CSF-1 eluted at approximately 120 mM sodium chloride.
  • Ammonium sulfate (4 M stock, pH 7.0) was then added to the 95- ml DEAE pool to a final concentration of 1 M.
  • the CSF-1 was then filtered through a Nalgene 0.45 micron filter and loaded (at 4°C) onto a 21.5 x 150 mm Bio-Rad TSK Phenyl-5-PW column equilibrated in depyrogenated 1.5 M ammonium sulfate, 0.1 M sodium phosphate (pH 7.0).
  • the column was washed witii two bed volumes of this loading buffer and tiien eluted in 0.1 M sodium phosphate (pH 7.0) using a 45-minute gradient in which the ammonium sulfate concentration decreased from 1.5 M to 0 M and die etiiylene glycol concentration increased from 0-60% (v/v).
  • the CSF-1 eluted at approximately 0.6 M ammonium sulfate in 30% ethylene glycol.
  • the CSF-1 was then extensively dialyzed into 10 mM Hepes buffer (pH 7.5) containing 150 mM sodium chloride and subsequendy was filter sterilized through a Millex 0.45 micron filter.
  • IL ____li protein produced under control of the P L promoter from DNA encoding SCSF/ V2CV150 as described in
  • European Patent Publication No. 0272779 was refolded and purified in a similar manner.
  • the vector used to transform the E. coli was prepared by substituting the appropriate synthetic fragment for the excised HmdHI/BstXI
  • the CSF-1 protein (SCSF/NV2C 150) in Section B was dialyzed (1 mg protein/ml) into a medium of 10 mM Hepes buffer, pH 7.2 containing 100 mM NaCl.
  • the PEG*-7000 was dissolved rapidly in a small volume of water and immediately added to the protein solution, which was continuously stirred throughout the reaction. A 3-fold to 30-fold molar excess of PEG*-7000 over CSF-1 dimer was employed. Reaction was complete in about 30 minutes at 20°C and in about three hours at 4°C.
  • FIG. 3A shows the chromatogram of underivatized rCSF-1 (SCSF/NV2CV150) sample prior to the experiment.
  • Figure 3B shows the chromatogram of 100 ⁇ g of the same CSF-1 sample after derivatization with a 5-fold molar excess ofPEG*-7000 for 30 minutes at 20°C. Radioimmunoassay, performed according to die method of Stanley, R. and Guilbert, J. Imm. Methods.
  • the fractions 30, 32, 34, 36, 37, 38, 39, 41 and 42 were also analyzed for biological activity, using the mouse bone marrow assay described by Moore et al., J. Immunol. (1983) 131:2397 and by Prystowsky et al., Am.
  • Biologically active protein was purified and refolded after expression of a construct encoding SCSF/CV150 in E. coli under control of the P promoter in a vector constructed as described in European Patent Publication No. 0272779 published June 29, 1988.
  • the strain used was an R coli ⁇ lysogen, DG116, transformed with the plasmid O/E pPi, CSF- 17 asp5 9 /CV150 (ATCC No. 67,389).
  • the protein was produced intracellularly in a monomeric, insoluble form, and purified and refolded as described in PCT Publication No. WO 88/08003 supra.
  • Figure 4 shows a size exclusion HPLC chromatogram of 8 mg of rCSF- 1 (SCSF/CV150) derivatized with PEG"-11,000.
  • the mildly derivatized fraction, pooled as shown in the figure, was found to retain essentially full biological activity, and to migrate with an apparent molecular weight of 80,000 daltons on sizing, and 45,000 daltons on nonreduced SDS-PAGE (Figure 8). This fraction was recovered at an overall yield of about 20%.
  • PEG-CSF The difference in sizes of PEG-CSF estimated by these two techniques is consistent with observations made by od ers.
  • PEG can alter the ability of a protein to bind SDS, affecting mobility on SDS-PAGE; and it can also affect size exclusion estimations, e.g., by hydrophobic interaction with d e column matrix.
  • LAL assay is described by a product brochure (1982) for PyroteE brand of LAL available from Associates of Cape Cod, Inc., Woods Hole, MA; it is also described by Watson et al. (eds.) "Endotoxins and Their Detection with the Limulus Amebocyte Lysate Test", Proceedings of an International Conference on Endotoxin Standards and Limulus Amebocyte Lysate Use with Parenteral Drugs. Alan R. Liss, Inc., New York (1982); and by Levin et al. (1964) Bull. Johns. Hopkins Hosp.. 115:265.
  • Figure 5 compares the time courses of blood clearance of modified and unmodified CSF-1 protein.
  • the systemic clearance is calculated by dividing die dose by die area under die blood plasma curve.
  • the data show tiiat the systemic clearance of derivatized rCSF-1 in d e blood is 0.302 ml/min kg versus 3.84 ml/min/kg for underivatized rCSF-1. This represents a 12.7-fold extended residence time of die derivatized as compared to die underivatized protein.
  • Figure 6 shows size exclusion HPLC of the rat blood plasma
  • Biochem, 112:195-203 developed with an antiserum to recombinant CSF-1 capable of detecting CSF-1 fragments, followed by ' - ⁇ I-p ⁇ otein A) of a non- reduced SDS-PAGE gel of urine and plasma verified tiiat the rCSF-1 antigen detected by Westem blotting was apparently intact, dimeric, and die same size as the material that was injected.
  • Sodium-naphthalene was prepared by addition of 0.15 g Na to a solution of 0.64 g naphthalene in about 20 ml tetrahydrofuran (THF) freshly distilled from sodium benzophenone.
  • THF tetrahydrofuran
  • Monometi ylpoly(ethylene glycol) of average molecular weight of 5000 (“mPEG 5000") was dried overnight in a vacuum dessicator with P 2 O 5 .
  • the Na-naptiialene solution was added dropwise to a solution of 2.5 g mPEG 5000 in -50 ml dry THF (freshly distilled). When the green color persisted in the solution to indicate excess, base addition was ceased and 1.2 ml BrCH 2 COOCH 3 was added dropwise.
  • a total of 1.5 mg of lyophilized residues was dissolved in 1.0 ml of 0.01 M phosphate, pH 7.0. The absorbance was 0.0680. A total of 50 ⁇ l of 5N NaOH was added and the A 400 was 1.106 (% ester - 93.9). The residues on the filter from the main precipitate were washed witii water and lyophilized over a weekend. The weight was 131 mg of fluffy white powder. A small amount was dissolved in 0.01 M phosphate, pH 7, and die A 400 was 0.0859. On adding 50 ⁇ l 5N NaOH, the A 400 was 0.6794 (87.4% ester).
  • the para-nitrophenyl ester just described was coupled with die asp5 9 SCSF/C 150 refolded protein described in Example I as follows: The rCSF-1 (200 ⁇ g at 1 mg ml) was dialyzed into 20 mM
  • reaction is carried out in cuvettes in a dual-beam Hewlett- Packard spectrophotometer, release of para-nitrophenol anion can be monitored, permitting the coupling reaction to be stopped reproducibly after a given amount of release has occurred.
  • the precipitate was collected on a coarse scintered glass filter. The precipitate was then stirred with about 400 ml methylene chloride for approximately 1 hour and tiien filtered and concentrated to about 100 ml. The PEG ester was precipitated again by addition of die solution to 1 liter ether. The precipitate was collected using a glass fiber filter and dried in a vacuum dessicator. The yield was 17.5 grams. The product was assayed for p-nitrophenyl carbonate content
  • CSF-1 was produced essentially as described in Example I. CSF-1 was concentrated to about 10 mg ml in 0.05 M Na borate, pH 9.0. 5 mg PEG-PNP obtained as a solid from Example HI Part A was added per ml of CSF-1. (approximately a 1:1 M ratio). The reaction was continued for 2 hrs at room temperature.
  • the refractile body suspension was then solubilized, refolded, and purified also as described in Example I.
  • the final specific activity was about 1.5 x 10 s units/mg in a CSF-1 bioassay performed on a CSF-1 dependent cell line.
  • the sample was concentrated to 6 ml by Amicon Centricon-30 centrifugation and purified by size exclusion HPLC in tiiree identical 2-ml-load runs.
  • the column used was Bio-Sil® TSK-250, 600 x 21.5 mm (BioRad), equilibrated in 0.2 M Na 2 HP ⁇ 4 /NaH 2 P0 buffer pH 7.0.
  • the A 28O peaks of mildly PEGylated protein in the three runs were pooled (pool 1, SEC) and concentrated to a final 2-ml volume, which was reinjected on the same column.
  • the active, PEGylated fractions were pooled and concentrated.
  • the final pool (pool 2, SEC), representing 15% of the starting material, consisted of 6 mg of CSF-1 that had approximately 100% of die initial bioactivity of d e unmodified CSF-1 and contained a major species of PEG-CSF-1 that migrated at about 45 K apparent M r on SDS-PAGE. Small amounts (about 10%) of unmodified CSF-1 and more highly modified CSF-1 remained in the pooled product Characterization of size exclusion chromatography (SEC) pools 1 and 2 is shown in Tables IV and V tables below:
  • Bioassay using Bioassay (using RIA (as described a CSF-1 dependent a CSF- ldependent above) ("units"/ cell line) (units/ cell line)(units/ Sample A ⁇ gp unit A ⁇ unit) mg
  • Example I The same conditions as described in Example I were used to estimate the average intravenous clearance rate in three rats of CSF-1 and PEG- CSF-1 described in Section A of this example, except that the doses for both PEGylated and non-PEGylated CSF-1 were 6 mg/kg.
  • Figure 7 shows the curve of relative concentration of CSF-1 from the blood of three rats versus time after intravenous injection.
  • the clearance was found to be about 0.63 ml/min/kg for PEGylated CSF-1 and 7.50 mVmin/kg for non-PEGylated CSF-1 (tiiree hours as opposed to five minutes). This represents about a 12-fold increase in average residence time in the blood for die PEGylated molecule.
  • CSF-1 (SCSF NV3CV158) was conjugated according to Example HI. After fractionation by (NH) 2 S0 4 as described in Part A, approximately 600 mg of CSF-1 having the molecular weight of CSF-1 dimer conjugated to one PEG was purified. This conjugate had a specific activity of about 6.2 X 10 7 U/mg. About 80 mg of a mixture of CSF-1 conjugates having 1, 2 or 3 PEGs linked was also recovered and had a specific activity of about 3.2 X 10 7 U/mg.
  • PEG-IL-2 was obtained by d e procedures described in commonly owned copending U.S. Serial No. filed on January
  • mice Groups of five mice were injected intraperitoneally (ip) on day -
  • a second in vivo tumor model using PEGylated CSF-1 to prevent metastases in the B 16 murine melanoma cell line is also useful.
  • mice 1-10 x 10 4 tumor cells, suspended in 0.2 ml of Ca+ 2 and Mg+2- free HBSS, are inoculated into unanesti etized mice in the lateral tail vein. From 14 to 21 days after tumor cell inoculation, the mice are sacrificed and necropsied. During necropsy, the lungs and brain are removed, rinsed in water, and weighed. The lungs are then fixed in Bouin's solution, and die number of surface tumor nodules per pair of lungs are determined witii the aid of a dissecting microscope.
  • Recombinant human CSF-1 PEGylated prepared according to Example HI and having a potency of 1-25 x 10 7 U/mg and pharmacologically acceptable endotoxin levels is used.
  • CSF-1 is freshly obtained prior to each experiment from frozen stocks and diluted immediately prior to injection in USP 0.9% saline.
  • PEGylated CSF-1 is delivered i.v., i.p. or s.c. on schedules which are dependent upon d e particular PEGylated CSF- 1 CSF-1 preparation used.
  • the dosing levels used range up to 5mg/kg.
  • CSF-1 at doses of up to 400 ⁇ g/kg, starting two days before infection with a sub-lethal dose of cytomegalovirus (CMV).
  • CMV cytomegalovirus
  • Dosing schedules are dependent upon the particular PEGylated CSF-1 preparation used.
  • Mice are sacrificed on the tiiird day after infection and d e extent of viral replication in target organs such as the spleen is evaluated by plaque assay. The results show that mice treated with PEGylated CSF-1 have significantly lowered organ viral titer compared to the saline-treated control mice, indicating that CMV infection is less severe in PEGylated CSF-1 treated mice.
  • PEGylated CSF-1 may be tested in a lethal murine CMV infection model in outbred CD- 1 mice (this is in contrast to the above experiment using sub-lethal doses of CMV, in which organ titers are monitored).
  • PEGylated CSF-1 is administered i.p., s.c. or i.v. to mice at doses up to 4 mg/kg (per mouse) starting up to 24 hours before viral challenge, there is a significant increase in survival as compared to saline- treated control.
  • PEGylated CSF-1 may be used alone or in combination witii another lymphokine in the treatment of viral infections in general, and in particular, may be beneficial in immunosuppressive viral infection such as acquired immune deficiency syndrome (AIDS).
  • AIDS acquired immune deficiency syndrome
  • Outbred CD- 1 mice are administered purified recombinant human CSF-1 PEGylated as described in Example HI, in amounts up to approximately 2 mg kg per dose on various dosing schedules which are dependent upon die PEGylated CSF-1 preparation used.
  • Total white blood cell count, neutrophil count monocyte count and lymphocyte counts are determined. Increases in any of tiiese parameters may be useful in clinical or veterinary medicine as a stimulus of granulocyte or monocyte production and an enhancer of white blood count
  • PEGylated CSF-1 is assayed for wound healing using animal models and protocols such as d e Goretex miniature wound healing model of Goodson and Hunt, 1982, J. Surg. Res..22:394, in which implanted Goretex tubes fill up with invading macrophages, fibroblasts and other connective tissue cells, and collagen and fibrin deposition. Healing is assessed by examining tube contents microscopically.
  • a second model is die incisional wound healing model of Eisenger, ei al., 1988, Proc. Nad. Acad. Sci.. USA. £5: 1937, in which wounds are observed visually and punch biopsies are taken to monitor healing, density of cells, and number of epidermal cell layers arising from hair follicles.
  • a third model is a serosal model such as die heat- injured testicular serosa of Fotev, ei al., 1987, J. Pathol..151:209, in which healing is assessed in fixed sections by degree of mesotiielial resurfacing of the injured site.
  • the teachings of each of these models are incorporated herein by reference.
  • PEGylated CSF-1 is applied to the site of the wound by soaking a nonadhesive surgical dressing in amounts up to 1,000,000 U/ml of PEGylated CSF-1 in saline under sterile conditions as described in the incisional wound healing model reference using epidermal cell derived factor (EDF) for topical wounds.
  • EDF epidermal cell derived factor
  • similar amounts of PEGylated CSF-1 are introduced into Goretex tubes at the time of implantation as described in Goodson and Hunt, ibid, or PEGylated CSF- 1 may also be incorporated into a slow-release matrix and applied at die site of the wound (in .
  • Goretex tubes, in or under dressings, or by injection in the serosal cavity) or PEGylated CSF-1 is administered systemically (i.v., i.p., or s.c.) at doses of up to 1,000 ⁇ g/kg/day.
  • the healing rate of each model is measured and tissue repair evaluated in the presence and in the absence of PEGylated CSF-1.
  • PEGylated CSF-1 may also be used in combination with other growth factors to promote wound healing such as EDF, epidermal growth factor (EGF), fibroblast growth factor (basic and acidic FGF), platelet derived growth factor (PDGF), or transforming growth factors (TGF alpha and beta), IL-1 and otiier substances such as somatomedin C and vitamin C.
  • EDF epidermal growth factor
  • fibroblast growth factor basic and acidic FGF
  • PDGF alpha and beta platelet derived growth factor
  • TGF alpha and beta transforming growth factors
  • LCSF/NV3CV221 was prepared according to fermentation purification and refolding procedures disclosed in PCT Publication No. WO88/08003 published October 20, 1988. 5 mg of LCSF was dialyzed into 20 mM HEPES buffer, pH 7.5, containing 50 mM NaCl. PEG"-11,000 was added at 6-fold molar excess over LCSF dimer. The dry activated PEG was added to die stirring 2 mg/ml LCSF solution at 20 ⁇ C. Aliquots of the reaction mixture were analyzed every 10 min by size exclusion HPLC (SEC) on Zorbax GF250 (Dupont). The reaction was stopped by addition of a 10-fold excess of ⁇ -aminocaproate over PEG. Table VHI shows NFS 60 bioassay data of die
  • Size exclusion analysis of the PEGylation reaction showed two major species ofPEG-CSF after the 30 min reaction. Their sizes were consistent with an approximate derivatization of 1 and 2 moles PEG per mole CSF-1 dimer. The SEC profile after 45 min indicated that die reaction had reached completion at about 30 min.
  • the sample was adjusted to pH 8.3 witii 1M Tris-HCl, diafiltered to educe salt concentration to 30 mM and applied to a Bio-Gel® TSK-DEAE-5-PW HPLC 75 x 7.5 mm column.
  • the column was equilibrated in 30 M Tris-HCl, pH8.5, and was developed widi a 40 min gradient of 0 to 0.6 M NaCl. All buffers were prepared with depyrogenated water, and die HPLC system was prewashed with 0.1 M NaOH and with 50% etiianol to remove endotoxin. Fractions off the column were analyzed by SDS-PAGE. PEGylated species eluted earlier than unmodified CSF-1 from the column. Some PEG-CSF also appeared in the column passthrough and this material again did not bind upon reapplication to the column. The unbound fraction also contained released NHS anion.
  • reaction herein may also be carried out employing polyvinyl alcohol or a polyoxyethylated polyol.
  • An active POG-CSF-1 may be prepared as follows.
  • Polyoxyethylated glycerol (POG) of molecular weight 5000 may be obtained from Polysciences. To 10 g of POG may be added 2.28 g glutaric anhydride (a 10-fold excess over POG). The mixture may be stirred for two hours at 110°C and cooled. This may be dissolved in 20 ml CHCI 3 and poured slowly into 500 ml ether with vigorous stirring. The product may be collected and rinsed witii ether to yield about 90% POG-glutarate product. This product may be reacted witii N-hydroxysuccinimide as described in Example IA to yield d e active ester POG-glutaryl N-hydroxysuccinimide (POG*). Then one of the CSF-1 proteins described above may be reacted with the POG*.
  • POG Polyoxyethylated glycerol
  • Nitrile-substituted polyvinyl alcohol may also be used to prepare an activated polyvinyl alcohol for conjugation to the CSF-1.
  • CMCC Cetus Master Culture Collection
  • ATCC American Type Culture Collection
  • the present invention is seen to provide various recombinant CSF-1 molecules selectively derivatized witii water-soluble polymers of different sizes using different chemical linkers.
  • Derivatization of the CSF-1 increases the apparent molecular weight of the CSF-1 and increases its in vivo half-life in the plasma of rats.
  • the derivatization may also increase the solubility of the CSF-1 in aqueous medium at physiological pH and may decrease its immunogenicity by decreasing or eliminating aggregation or by shielding its antigenic determinants.

Abstract

A biologically active CSF-1 protein is selectively conjugated via certain amino acid residues or carbohydrate moieties to a water-soluble polymer selected from polyethylene glycol or polypropylene glycol homopolymers, polyoxyethylated polyols, or polyvinyl alcohol. The resulting conjugated CSF-1 is biologically active and has increased circulating half-life in mammals, compared to that of the unconjugated protein. The conjugated CSF-1 may be used to stimulate the immune response or to provide more cells to be stimulated.

Description

CONJUGATION OF POLYMER TO COLONY STIMULATING FACTOR- 1
This invention relates to a chemical modification of biologically active colony stimulating factor- 1 (CSF-1) that alters the chemical and/or physiological properties of this protein. More specifically, this invention relates to selective conjugation of CSF-1 to polymers to increase the circulating half-life of the protein in mammals.
Colony stimulating factor- 1 (CSF-1) (also known as M-CSF) is one of several proteins that are capable of stimulating colony formation by bone marrow cells plated in semisolid culture medium. CSF-1 is distinguished from other colony stimulating factors by its ability to stimulate the formation of predominantly macrophage colonies. Other CSFs stimulate the production of colonies that consist of neutrophilic granulocytes and macrophages, exclusively neutrophilic granulocytes, or neutrophilic and eosinophilic granulocytes and macrophages. A review of these CSFs has been published by Dexter, T.M., Nature (1984) 202:746, and by Vadas, M.A.. J. Immunol (1983) 130:793. There is currendy no routine in vivo assay that is known to be specific for CSF-1 activity.
CSF-1 has been purified from native sources (see, e.g., Csejtey et al., Biochem. Biophvs. Res. Comm. (1986) 12&238 and PCT publication No. WO 86/04587 published August 14, 1986 regarding immunoaffinity chromatography of native CSF-1 to enable partial amino acid determinations). CSF-1 has also been produced from recombinant DNA using two apparendy related cDNA clones: (1) a "short" form that encodes a monomeric protein of 224 amino acids preceded by a 32-amino acid signal sequence (Kawasaki et al., Science (1985) 220:292-296); and (2) a "long" form, encoding a monomeric protein of 522 amino acids, also preceded by the 32-amino acid signal sequence. The long form has been cloned and expressed by two groups, as disclosed in European Patent Publication No. 0272779 published June 29, 1988 and Ladner et al. EMBO J. (1987) _5:2693, each incorporated herein by reference; and in Wong et al. Science (1987) 235:1504- 1509, and PCT WO87/06954 published November 19, 1987. (The DNA and amino acid sequences for these two clones are shown in Figures 1 and 2, respectively.) Both the long and short forms of CSF-1 are described by Clark and Kamen, Science (1987) 22_5: 1229- 1237. An "intermediate" form that encodes a monomeric protein of 406 amino acids preceded by a 32-amino acid signal sequence has also been recently reported (Cerretti et al. (1988) Mol. Immunol. 22:761-770).
The long and short forms of the CSF-1-encoding DNA appear to arise from a variable splice junction at the upstream portion of exon 6 of the genomic CSF-1-encoding DNA. When CSF-1 is expressed in certain eucaryotic cells from either the long or short cDNA forms, it is secreted as a dimeric glycoprotein and appears to be variably processed at the C-terminus and/or variably glycosylated. Consequently, CSF-1 proteins of varying molecular weights are found when the reduced monomeric form is subjected to Western analysis.
The amino acid sequences of the long and short forms, as predicted from the DNA sequence of the isolated clones and by their relationship to the genomic sequence, are identical in the first 149 amino acids at the N-terminus after signal peptide cleavage, and diverge thereafter as a result of the insertion in the longer clone of an additional 894 bp fragment (encoding 298 additional amino acids) before the codon encoding amino acid 150. Therefore, both the shorter and longer forms of the gene encode regions of identical sequence at the C-terminus, as well as at the N-terminus. Biologically active protein has been recovered when truncated cDNAs encoding only the first 145 or 147 amino acids of the mature short form (European -Patent Publication No.0261592 published March 30, 1988; Cerretti et al. supra or the first 190 or 221 amino acids of the mature longer form, are expressed in eucaryotic cells.
Recombinant CSF-1 was expressed in I £c_U by modifying a short clone cDNA originally described by Kawasaki et al., Science (1985) 230:291 to code for proteins that contained 1) the native N-terminus and a C- terminus at amino acid 150 of the mature protein, and 2) a truncation to delete the first two amino acids at the N-terminus and a C-terminus at amino acid 150 of the mature protein. These proteins were purified and refolded to form homodimers and were found to have apparent molecular weights on size- exclusion high performance liquid chromatography (HPLC) of about 43,000 and 40,000 daltons, respectively. CSF-1 proteins modified so that the C- terminus of the expressed protein is amino acid 150 or 158 and so that up to three amino acids at the N-terminus are deleted have also been prepared. Small proteins (less than about 70 kd) often have a relatively short half-life in blood after intravenous injection. Rapid clearance of drugs from circulation often reduces their efficacy. It is often desirable to increase the half-life of a circulating polypeptide so that smaller amounts of the polypeptide or less frequent injections might be administered, while retaining the desired therapeutic effect Modifications of the CSF-1 protein that might alter its half-life in vivo, reduce its immunogenicity, or reduce or eliminate aggregation of the protein that might occur when it is introduced in vivo would be desirable. Such modifications include the modification of proteins with substantially straight chain polymers such as polyethylene glycol (PEG), polypropylene glycol (PPG), dextran, or polyvinyl alcohol. For example, U.S. Patent No. 4,261,973 describes conjugation of immunogenic allergen molecules with non-immunogenic water- soluble polymers such as PEG or polyvinyl alcohol to reduce the immunogenicity of the allergen. U.S. Patent No. 4,301,144 describes conjugation of hemoglobin to PEG, PPG, a copolymer of ethylene glycol with propylene glycol, or ethers, esters or dehydrated products of such polymers to increase the oxygen-carrying ability of the hemoglobin molecule. U.S. Patent No.4,609,546 discloses that conjugation of a polypeptide or glycoprotein such as a colony stimulating factor to a polyoxyethylene-polyoxypropylene copolymer may increase the duration of its physiological activity. The only proteins that have been tested in this fashion are enzymes or native interferon, which are readily water-soluble. PCT WO 86/04145 published July 17, 1986 discloses PEG modification of antibodies to decrease binding to Fc receptors. U.S. Patent No. 4,179,337 discloses conjugation of water-soluble polypeptides such as enzymes and insulin to PEG or PPG to reduce the immunogenicity of the polypeptides while retaining a substantial proportion of their desired physiological activities. EP 154,316, published September 11, 1985 to Takeda Chemical Industries, Ltd., discloses and claims chemically modified lymphokines such as IL-2 containing PEG bonded directly to at least one primary amino group of the lymphokine. In addition, Katre et al., Proc. Natl. Acad. Sci. (1987) 8_4: 1487 discloses modification of IL-2 with PEG. Many other references disclose the concept of PEG derivatization of proteins such as alpha- 1-proteinase inhibitor, asparaginase, uricase, superoxide dismutase, streptokinase, plasminogen activator, IgG, albumin, lipoprotein lipase, horseradish peroxidase, catalase, arginase and asparaginase, as well as peptides. Such derivatization through lysines was reported as improving half-life, decreasing immunogenicity, increasing solubility, and in general, increasing efficacy (which permitted less frequent dosing). In most cases, the proteins required multiple modifications per molecule to achieve improved performance jn vivo, and the activity in vitro was significandy decreased by such modification. Modification of IL-2, IFN-β and immunotoxins with PEG through cysteine residues of a polypeptide is disclosed in PCT WO87/00056 published January 15, 1987.
Li addition to these patents and patent publications, several articles discuss the concept of using activated PEG or PPG as a modifying agent for proteins such as enzymes, IgG and albumin. For example, Inada et al., Biochem. and Biophvs. Res. Comm.. 122. 845-850 (1984) disclose modifying water-soluble lipoprotein lipase to make it soluble in organic solvents such as benzene by using cyanuric chloride to conjugate with PEG. Takahashi et al., Biochem. and Biophvs. Res. Comm.. 121:261-265 (1984) disclose modifying horseradish peroxidase using cyanuric chloride triazine with PEG to make the water-soluble enzyme active and soluble in benzene. Patents and patent publications that disclose use of polyvinyl alcohol (PVA) in protein conjugation reactions include U.S. Patent Nos. 4,296,097 and 4,430,260, relating to conjugation of benzylpenicilliri and PVA, U.S. Patent No. 4,496,689 (EP 147,761), relating to conjugation of alpha-1-proteinase inhibitor with a polymer such as heparin, PVA or PEG, EP 142,125 published May 22, 1985, disclosing non-covalent bonding of hemoglobin to PVA as a carrier, DE 2312615 (Exploaterings AB TBF), relating to crosslinked, water-insoluble PVA coupled to a protein, and DE 3,340,592 published May 23, 1985, relating to conjugates of PVA with human hemoglobin A.
Articles relating to conjugates of proteins and PVA include Sabet et al., Indian J. Chem.. Sec. A (1984) 23A(5) (disclosing PVA and protein interaction), Wei et al., Immunol. (1984) 5I(4):687-696 (disclosing trimellityl conjugated with PVA), Lee et al., J. Immunol. (1981) 126:414-418 and Hubbard et al., J. Immunol. (1981) 126:407-413 (both disclosing DNP conjugated to PVA), Lee et al., Int. Arch. Allergy Appl. Immunol. (1980) 62:1-13 (disclosing antibenzylpenicilloyl IgE conjugated to PVA), Sehon, Pro . Allergy (1982) 22:161-202 (disclosing an allergen and hapten conjugated via PVA), Holford-Strevens et al., Int. Arch. Allergy App. Immunol. (1982) 67:109-116 (disclosing conjugation of PVA and an antigen/hapten), and Sehon and Lee, Int. Arch. Allergy App. Immunol. (1981) 66 (Supp. 1), p. 39-42 (disclosing a hapten/allergen conjugated to PVA).
PCT Publication Nos. WO 86/04607 and WO 87/03204 and Ralph et al., Immunobiol. (1986) 172:194 disclose various potential uses of CSF-1, including use as an anti-infection, anti-tumor, or wound-healing agent.
None of these references, however, discloses details of how to modify CSF-1 with a polymer such as PEG or polyvinyl alcohol so as to retain its biological activity while also increasing its circulating half-life or efficacy. Furthermore, it is not generally possible to predict the extent of protein modification or the nature of the reaction conditions that are desirable, because some proteins are much more susceptible to inactivation through conjugation than others. Accordingly, the present invention provides for modifying colony stimulating factor- 1 to increase its half-life in vivo, while also retaining useful biological activity. The modified CSF-1 may be used to stimulate cells In vivo at much reduced and/or less frequent dosage than the unmodified CSF- 1. As secondary advantages, the modification may decrease the immunogenicity of the CSF-1 (especially when used in heterologous species, such as use of human CSF-1 for cattle) and/or reduce aggregation of the protein that might occur.
More specifically, the present invention is directed to a biologically active composition having prolonged in vivo half-life in mammals, comprising a protein that stimulates the formation of primarily macrophage colonies in the in vitro colony stimulating factor- 1 assay, which protein is covalendy conjugated to a water-soluble polymer selected from the group consisting of polyethylene glycol or polypropylene glycol homopolymers, polyoxyethylated polyols, and polyvinyl alcohol, wherein said homopolymer is unsubstituted or substituted at one end with an alkyl group. The CSF-1 protein may be conjugated to the polymer via: (1) free amino groups, including lysine residues and the N-terminal amino acid (preferably 1 to 3 sites), (2) carbohydrate moiety(ies) of eukaryote-expressed, glycosylated CSF-1; or (3) free sulfhydryl groups engineered into the CSF-1 Preferably the polymer is unsubstituted polyethylene glycol (PEG), monomethyl PEG (mPEG), or polyoxyethylated glycerol (POG) that is coupled to the (1) lysine residue(s) of the CSF-1 via an amide or urethane linkage formed from an active ester (preferably the N-hydroxysucάnimide or paranitrophenyl ester) of a PEG, mPEG, or POG carboxylic or carbonic acid; (2) carbohydrate moiety(ies) of the CSF-1 via an amine, hydrazine, or hydrazide linkage; or (3) cysteine residue(s) of the CSF-1 via a maleimido or haloacetyl (where halo is Br, Cl or I) group..
Another aspect of this invention resides in a process for preparing the conjugated protein described above, comprising:
(a) providing a water-soluble polymer having at least one terminal reactive group where the polymer is selected from the group consisting of polyethylene or polypropylene glycol homopolymers and polyoxyethylated polyols, and polyvinyl alcohol, wherein said homopolymer is unsubstituted or substituted at one end with an alkyl group;
(b) reacting the protein with the reactive group of said polymer so as to render the protein biologically active and selectively conjugated; and
(c) purifying the conjugated protein.
In another aspect, the invention relates to a method for prophylactic or therapeutic treatment of infectious diseases or cancer in mammals and to a methods effective for cancer treatment, osteroporesis treatment, wound healing, cholesterol lowering or antibody dependent cellular cytotoxicity (ADCC) in mammals comprising administering to the mammal an effective amount of a pharmaceutical preparation comprising the conjugated CSF-1 protein dissolved in a pharmaceutically acceptable aqueous carrier medium.
Figure 1 shows the cDNA and deduced amino acid sequence for pCSF-17 (32 amino acid leader sequence and amino acids 1-224).
Figure 2 shows the cDNA and deduced amino acid sequence for CSF-4 (partial amino acid leader sequence and amino acids 1-522).
Figure 3A shows the size exclusion HPLC chromatogram of underivatized recombinant CSF-1 (rCSF-1) (SCSF/NV2CV150). Figure 3B shows the HPLC chromatogram of the same rCSF-1 derivatized with PEG- NHS of 7000 dalton average molecular weight. Figure 4 shows a size exclusion HPLC chromatogram of rCSF- 1 (SCSF/CV150) derivatized with PEG-NHS of 11,000 daltons average molecular weight.
Figure 5 shows a graph of CSF-1 concentration in rat blood plasma versus time for R gρji rCSF- 1 (SCSF/CV150), the dimeric product derived from the rCSF-1 with the sequence in Figure 2 [lacking the leader sequence and a C-terminal sequence, but retaining essentially all of the N- terminal methionine present in the E. cρji construct], rCSF-1 expressed in mammalian cells (COS) that arises from a glycosylated 522-amino acid precursor (LCSF) andPEG-ll.OOO-derivatized R coH rCSF-l (SCSF/CV150).
Figure 6 shows a size exclusion HPLC chromatogram of rat blood plasma 120 minutes after intravenous injection of PEG- 11,000- deτivatizedrCSF-1 (SCSF/CV150). Radioimmunoassay (RIA) and absorbance at 280 nm are plotted.
Figure 7 shows a graph of CSF-1 concentration in rat blood plasma versus time for K coti rCSF-1 (SCSF/NV3C 158) and for the same protein derivatized with 11,000 PEG.
Figure 8 shows SDS-PAGE analysis of rCSF-1 derivatized with PEG-11,000 (SCSF/CV150). Gel (10%), stained for protein with
Coomassie blue, is of the sample used in Figure 5, with and without reduction of disulfide bonds.
Definitions
"Colony stimulating factor- 1" refers to a dimeric protein or glycoprotein composition that has the biological activity for CSF-1 in the standard in vitro colony stimulating assay of Metcalf. J. Cell. Physiol. (1970) 26:89 wherein the polypeptide stimulates the formation of primarily macrophage colonies. "Biologically active CSF-1" means a composition of conjugated CSF-1, that has essentially the same specific activity in mouse bone marrow colony-forming assays as the unconjugated form of the same CSF-1 or at least about 10% of its specific activity. A "native" CSF-1 is a non- recombinant CSF-1. Murine CSF-1 is described in European Patent Publication No. 0272779, Rajavashisth et al., Proc. Natl. Acad. Sci. USA (1987) Mil 157 and in DeLamarter et al., Nucleic Acids Res. (1987) 11:2389.
"Clinically pure" CSF-1 means a preparation of biologically active human CSF-1 produced recombinantly in bacteria that is at least 95% CSF-1 either by RP-HPLC or by either reducing or non-reducing SDS-PAGE analysis and has an endotoxin content of less than about 1.0 ng/mg CSF-1.
"Selectively conjugated" refers to proteins that are covalently bonded via free amino groups of the protein (preferably one or two free amino groups, to retain maximum biological activity), via free sulfhydryl groups (if any), or via a carbohydrate moiety (if any) present in the protein. "Effective amount" and "immunotherapeutically effective amount" signify an amount effective to perform the function specified, such as to promote tumor reduction or prevent or cure infectious diseases. The exact optimal amount will depend on many factors, including the patient's clinical history and current disease, die schedule, the route, and the response of the patient
"Therapeutic treatment" indicates treating after the disease is contracted, whereas "prophylactic" treatment indicates treating to prevent contraction of the disease.
"Mammals" indicates any mammalian species, and includes rabbits, mice, dogs, cats, cattle, sheep, primates, and humans, preferably humans.
"Muteins" are genetically engineered proteins expressed from a nucleic acid sequence that has been altered using techniques such as site- specific mutagenesis. Such genetic alterations are designed to result in one or more substitutions, additions, and/or deletions to the amino acid sequence of the parent protein.
"Pharmaceutically acceptable" refers to a carrier medium that does not interfere with the effectiveness of the biological activity of the active ingredient(s), is stable, and is not toxic to the host to whom it is administered. "Homodimer" refers to a dimeric protein essentially identical in its two subunits except that it also includes dimeric proteins with minor microheterogeneities that occasionally arise on production or processing of recombinant proteins.
'Ηeterodimer" refers to a dimeric protein having subunits which differ in amino acid sequence, the number of amino acids or both. In other words, heterodimers have two nonidentical subunits. CSF-1 Proteins .
As set forth in the background section, CSF-1 is biologically active in its dimeric form. The CSF-1 employed herein may be the native dimer or recombinantly produced dimer. Native dimeric CSF-1 species have been obtained from human urine, cultured monocytes, and culture supematants of some cell lines. It has been possible to obtain recombinant DNA encoding CSF-1 monomers consisting of a variety of amino acid sequences and lengths. Figures 1 and 2 show the DNA sequences and amino acid sequences predicted from the DNA sequences for, respectively, the full- length, unprocessed short and long forms, both of which contain a 32-amino acid signal sequence at their N-termini. The sequences of the monomeric CSF- 1 protein are considered herein for convenience to be the 224-amino-acid short form without the leader sequence (designated herein as SCSF), and the 522- amino-acid long form without the leader sequence (designated herein as LCSF). Other CSF-1 dimers produced from DNA sequences modified by point mutation(s), insertion(s), deletion(s), and/or translocation(s) are also expected to benefit from the chemical modification of this invention if they are biologically active.
The recombinant CSF-1, produced in any host, whether eukaryotic or prokaryotic, may be conjugated to polymers via selected amino acid side groups, preferably free amino groups. Preferably, the DNA encoding CSF-1 is expressed in bacteria and the resulting CSF-1 is a homodimer after purification and refolding. If the conjugation is via a carbohydrate moiety, the host may be eukaryotic, or glycosylation may be carried out in vitro.
For convenience, the primary structure of the protein subunits encoded by the various cDNA constructs described will be designated herein using a shorthand notation, as follows: LCSF denotes the 522-amino acid sequence disclosed for the clone CSF-4, set forth in European Patent Publication No. 0272779 referred to above and shown in Figure 2, without the complete signal sequence. SCSF denotes the 224-amino acid sequence disclosed for the clone pCSF-17, shown in Figure 1, without the signal sequence, described in the Kawasaki article referred to above, Science (1985) 220:292-296, also incorporated herein by reference. It will be noted that one particular pCSF-17 clone derivative has an aspartic acid residue at position 59. The disclosed LCSF clone also encodes Asp at position 59. Muteins corresponding to amino acid substitutions within the sequences depicted in Figures 1 and 2 are correspondingly designated by the substitution subscripted with the position. Mutein forms of CSF-1 are disclosed in European Patent Publication No. 0272779 published June 29, 1988, the disclosure of which is incorporated herein by reference. When constructs encoding these proteins are expressed in bacteria, the final products may also retain N-terminal methionine to varying degrees. Since the extent of N-terminal methionine removal cannot be reliably predicted, this possibility is not included in the notation.
C-terminal and N-terminal truncations of these basic SCSF and
LCSF sequences will be designated as CV or NV, respectively. The C- terminal deletions will be followed by the number representing the number of amino acids of the native structure remaining; for the -N-terminal deletions,
NV will be followed by the number of amino acids deleted from the N- terminus. Thus, for example, LCSF/C 150 denotes a construct encoding a protein that contains the first 150 amino acid residues of the long CSF sequence; SCSF/CV158 denotes a construct encoding a protein that contains the first 158 amino acid residues of the short form; SCSF/NV2 denotes a construct encoding the short form with two N-terminal amino acids deleted. (As set forth above, the LCSF and SCSF diverge beginning at position 150 andreconverge after 298 amino acids in the LCSF clone.) LCSF/NV2CV150 denotes a form that is the same as LCSF/CV150, except that the two N- terminal glutamic acid residues are deleted.
Plasmids encoding a variety of CSF-1 forms are currently available, and can be expressed in bacterial systems. A form of plasmid encoding the long form of CSF-1 can be expressed in eukaryotic cells, in which case, the eukaryotic cell "processes" the clone, secreting a protein that is C-terminally truncated and has the leader sequence removed from the N- terminus. Alternatively, the clone can be truncated to express specific C- terminally deleted forms in eukaryotic or prokaryotic cells. In addition, the first two or three N-terminal codons can be deleted so that the resulting protein expressed in E. coli is more homogeneous. Specifically, the N-terminal methionine encoded just upstream of the mature native sequence N-terminus in the E. coli constructs (which is retained in the protein as "N-terminal Met" unless removed by post-translational processing) is more readily removed from these N-terminal deletion constructs. Furthermore, significant heterogeneity detectable using reverse-phase HPLC (RP-HPLC) is found when the gene encoding the "native" N-terminal sequence (for example, of the short form, mutein SCSF/CV150) is expressed in E. coli and the purified product is characterized. This heterogeneity is ehminated when the corresponding CSF-1 gene lacking the two N-terminal codons (glutamic acid) is expressed. Correspondingly, N-terminal truncated forms of other short and long CSF-1 gene constructs can also be employed.
Preferred constructions are those wherein the protein consists essentially of an amino acid sequence selected from the group consisting of
LCSF/CV150 through CV464, tyr59LCSF/C 150 through CV464; asp5-SCSF/CV150 through CV166. More preferred are those CSF-1 proteins consisting essentially of an amino acid sequence selected from the group consisting of LCSF/CV150; LCSF/CV190; LCSF/CV221;
LCSF/CV223; LCSF/CV236; LCSF/CV238; LCSF/CV249;
LCSF/CV258; LCSF/CV406; LCSF/CV411; LCSF/CV464; asp59SCSF/CV150; asp5. SCSF/CV153; and asp5oSCSF/CVl58.
Particular preferred constructions that result in CSF-1 proteins for modification herein include clones encoding LCSF/CV190,
LCSF/CV221, asp59SCSF/CV158, asp59SCSF/CV150 and their corresponding NV2 and NV3 forms. The most preferred starting materials are the products of the clones encoding asp59SCSF/CV150; asp59SCSF/CVl58; LCSF/CV221 and their corresponding NV3 forms.
Alternatively, the CSF-1 may be in the form of a heterodimer prepared from among various monomeric units of CSF-1, particularly if the conjugation is done through a reactive cysteine residue of the CSF-1 heterodimer. The large number of CSF-1 proteins formed by variations in C- terminal sequence due either to differences in processing or clone construction provides a variety of starting materials that can be utilized in heterodimer formation. Thus, novel heterodimeric materials can be formed by refolding.
For example, the monomeric form of SCSF/CV150, along with the monomeric form of LCSF/CV157, can be mixed and treated according to the method of PCT Publiction No. WO 88/08003 published October 20, 1988, the disclosure of which is incorporated herein by reference. The heterodimer can then be separated from the homodimeric and oligomeric side products by various chromatographic and other methods. The heterodimer (particularly
SCSF/CV150 and LCSF/CV157) can be made to terminate on one subunit at the 157 position, providing a potentially free sulfhydryl group for reaction with the conjugation polymer. Similar mixtures subjected to the method of the invention could lead to heterodimers of components having amino acid substitutions~e.g., glu52 LCSF/CV221 andLCSF/C 190.
The differing monomers may be mixed in vitro or produced in the same cell. If produced in the same cell, a construct for expression of each monomer is introduced into the same host; in such embodiments, it is preferred that each construct bear a different marker (such as tetracycline resistance (TcR) and Ampicillin resistance (AmpR), so that cotransformed hosts are selected. The cotransformed cells are then grown and induced to obtain mixtures of the two forms.
In addition, single cysteine residues can be engineered into CSF-1 in a non-natural location to create a construct of rCSF-1 containing a free sulfhydryl group on a cysteine residue for PEG reaction, provided the molecule retains bioactivity. Heterodimer constructs containing a substitution of a given cysteine in only one of the two subunits may also be useful in the generation of free sulfhydryls. Also, carbohydrate moieties can be placed on the CSF-1 protein, either by expression from eukaryotic systems or by in vitro glycosylation with enzymes.
Recombinant CSF-1 proteins may or may not retain the exact length prescribed by the clone, due to processing by the host Therefore, although the starting material proteins are referred to by the same designation, it should be understood that these designations, in reality, refer to the clone construct and the actual length of the starting material for the process disclosed herein may be shorter, or longer (if it has N-terminal Met), than that specified by the C-terminal amino acid number. As mentioned above, the CSF-1 may be produced in eukaryotic cells, e.g., mammalian, insect, or yeast cells. Production in mammalian cells is described in PCT Patent Publication No. WO 86/04607. The CSF-1 may be produced from insect cells using a baculovirus vector, as described in Luckow et al., Biotechnol. (1988) 6:47, the disclosure of which is incorporated herein by reference. Other descriptions of this type of CSF-1 production are contained within Weaver et al., BioTechnologv (1988) 6:287.
Insect-cell-produced CSF-1 (SCSF/CV150) is a glycosylated dimer that was found to have a very short in vivo half-life. The two N-linked glycosylation signals in the amino acid sequence of SCSF (Asn 122 and Asn 140) could be modified in the DNA using site-directed mutagenesis to generate a non- glycosylated, bioactive insect-cell-produced CSF-1 protein (Gin 122, Gin 140). This protein can then be reacted via its lysine or cysteine residues with a polymer as done with prokaryote-expressed rCSF-1 to increase its in vivo half- life.
The polymer can be attached covalently to the glycosylated CSF-1 on the carbohydrate, by reacting, for example, PEG-amine, PEG- hydrazine, or PEG-hydrazide with CSF-1 protein that has been oxidized with periodate to convert vicinal diols of the sugars to aldehydes. The PEG-amine can be prepared by converting PEG-OH to PEG-tosylate and then to PEG- phthalimide, and the phthalimide is cleaved with hydrazine to produce PEG- NH2 in a Gabriel synthesis. PEG-hydrazine and PEG-hydrazide can be prepared by methods known to those skilled in the art. The PEG derivative will couple at carbohydrate sites where vicinal diols were converted to aldehydes.
If CSF-1 is produced intracellularly in bacterial hosts, it must be refolded to form active dimers in high yield, as described in PCT Publication No. WO 88/08003 supra. Briefly, in this procedure the solubilized monomer, whether secreted or produced as a mature or fusion protein, is maintained in a chaotropic environment under reducing conditions. Such maintenance involves the use of a suitable reducing agent such as β- mercaptoethanol or dithiothreitol (DTT). The solubilized protein is typically maintained in, for example, 8 M urea or 7 M guanidinium hydrochloride, at a pH of about 7-8.6, in the presence of about 2-100 mM thiol compound. Starting with this solubilized form, the monomer may either be refolded directly or purified from remaining proteins by a suitable purification procedure such as chromatography on an adsorbant gel, chromatography using an ion exchange column, or gel-permeation chromatography prior to refolding. Gel-permeation chromatography is useful, as it permits an easy size separation of the desired monomer length, which is generally known in advance, from impurities of differing molecular weights. DEAE Sepharose chromatography in urea is even more useful, as it concentrates as well as purifies and is more easily scaled up than is sizing. It is preferable that the purification be conducted under reducing conditions to prevent the formation of disulfide-linked aggregates. Thus, regardless of the chromatographic procedure used, a suitable reducing agent is included in the solutions used to load the chromatographic columns or batches and in the eluting solutions. In some instances, low pH may be substituted for reducing agent, as low pH will essentially prevent disulfide bond formation and is compatible with some chromatographic systems. In addition, inclusion of a low concentration of a chelating agent may help to preserve the reduced form of the protein.
The partially purified monomer is then subjected to refolding conditions for the formation of the dimer. The protein concentration during this step is of considerable importance. Generally, final percent yields per volume of the CSF-1 refolding reaction are increased if the protein concentration is less than about 2 mg ml of the CSF-1 protein; a concentration range of 0.03-1.0 mg/ml is preferred. The refolding conditions may include gradual removal of the chaotropic environment over an appropriate time period, usually several hours, or dilution of the sample to the desired concentration of protein and chaotropic agent. Also possible are methods that provide a constant protein concentration, such as dialysis or hollow fiber diafiltration, while the chaotrope is slowly removed. At the end of the process, when the chaotrope is depleted, a relatively nondenaturing level is reached. For example, if guanidine hydrochloride is used as a chaotrope, a final concentration of less than about 2 M, and preferably 0.1-1 M, is attained, and if urea is used as the chaotrope, a final concentration of less than about 1 M, and preferably 0.1-0.5 M, is attained.
The refolding is conducted so as to permit oxidation of the sulfhydryl groups to disulfides that establish the biologically active dimeric configuration that, in the case of CSF-1, requires the formation of disulfide bonds, one or more of which links the two chains. One or more intrachain disulfides are also formed. Suitable redox conditions that encourage this formation of dimers include the sulfhydryl/disulfide reagent combinations, such as oxidized and reduced glutathione. The ratio of reduced to oxidized glutathione or other sulfhydryl/disulfide combination is typically from about 2 mM/0.1 mM to 0.5 mM/1.0 mM. Other methods of oxidation are also acceptable. In any event the pH of the solution during the refolding process should be maintained at about 7.5 to 9.0 to favor the reaction. It is clear that during the process of refolding die highly reducing conditions under which the initial purification was conducted are no longer employed. The exclusion of significant concentrations of salts, such as sodium chloride, during the refolding process, permits the use of ion exchange chromatography as a subsequent concentration/purification step.
During the refolding process higher oligomeric species of CSF- 1 may be formed. This aggregation processes minimized through temperature control, wherein low temperatures of about 0-4°C are preferable to higher temperatures of 25-37°C.
Residual redox reagents present in die refolded CSF-1 preparation may possibly facilitate disulfide exchanges during subsequent purification steps. Two more preferred procedures to block such unwanted disulfide exchanges include lowering the pH to below 7.0 or diafiltration to remove the redox reagents.
For example, prior to further purification of the refolded, dimeric CSF-1, removal of the redox material and concentration of the refolded proteins may be performed by direct loading of the refolded material onto an ion exchange chromatography column using, for example, DEAE Sepharose. Frequendy such procedures are carried out at pHs around 8; however, lowering the pH into the range of 5.5 to 7.0 reduced oligomer formation and increased yield of dimeric CSF-1.
After refolding, concentration, and purification, the dimer is further purified from residual redox material and from other proteins using procedures similar to those set forth above for the monomer. Suitable means, in particular, include gel filtration, hydrophobic interaction chromatography, ion exchange chromatography, and reverse-phase HPLC.
A particularly successful protocol for removal of undesirable impurities such as pyrogens or other endotoxins includes the use of chromatography on a phenyl-TSK or phenyl-Sepharose column. The chromatography is carried out under conditions and with reagents that are essentially endotoxin-free. The desired dimeric CSF-1 is soluble and stable in approximately 1.5 M ammonium sulfate at neutral pH, and is loaded onto d e columns under these conditions at low temperatures, i.e., from about 4°C to about 10°C, and preferably about 4°C. Removing the precipitate that forms on adding die ammonium sulfate removes some aggregates and unstable forms of refolded CSF-1. The CSF-1 protein can be eluted using a gradient of decreasing ammonium sulfate (1.5 to 0 M) with increasing ethylene glycol (0 to 50%) in neutral buffer. The CSF-1 dimer elutes at approximately 0.6 M ammonium sulfate, 35% ethylene glycol from the phenyl-TSK column. Propylene glycol may be used instead of ethylene glycol, in which case the elution conditions will be somewhat different Alternative supports can also be used, andphenyl-Sepharose is, in fact preferred for larger scale production of die purified CSF-1 dimeric protein.
Conjugation
The CSF-1 protein described above is conjugated to die polymer via either (1) free amino group(s), preferably only one or two in order to minimize loss of biological activity, (2) at least one carbohydrate moiety on the protein, or (3) free sulfhydryl group(s) that is/are engineered into d e clone and remain free after refolding.
The number of polymer molecules tiiat have been conjugated to the protein can be determined by various methods, including, for example, acid degradation or digestion, followed by amino acid analysis if the links are maleimido or bromoacetyl to cysteine links and die extent of derivatization is high (more than 4 moles/mole). Altematively, the conjugated protein can be digested into small fragments with an enzyme (e.g., trypsin) and separated by column chromatography. A peptide map of d e protein before and after modification would be compared, and fragments with altered elution times sequenced to determine the location(s) of polymer attachments. Li a third alternative, the polymer can be radioactively labeled prior to coupling to determine how many moles of radioactive polymer are attached per mole of CSF-1 protein. i cases where polymers of relatively uniform molecular weight are conjugated to CSF-1 of uniform molecular weight measurement of die molecular weight of the conjugate can serve as an estimate of me number of polymers per CSF-1 molecule. The residue(s) to be conjugated may be: (1) any free amino groups (ε-amino groups at lysine residues or a free amine group, if any, at the
N-terminus), (2) free sulfhydryl groups on cysteine residues that are engineered or constructed into CSF-1, or (3) carbohydrate moiety (discussed elsewhere). It has been found tiiat if the protein is moderately derivatized on free amino groups witii PEG (i.e., contains about one to three modified amino acid residues), it retains from 25-100% of the bioactivity of underivatized CSF-1. If, however, it is highly derivatized witii PEG, the conjugated protein has significandy less measurable bioactivity, depending on die type of CSF-1, length of PEG polymer and the linker and reaction conditions employed.
The polymer to which the protein is attached is a homopolymer of polyethylene glycol (PEG) or of polypropylene glycol (PPG), a polyoxyethylated polyol, or polyvinyl alcohol, provided in all cases that the polymer is soluble in water at room temperature. Examples of polyoxyediylated polyols include polyoxyethylated glycerol, polyoxyethylated sorbitol, polyoxyediylated glucose, and die like.
The glycerol backbone of polyoxyethylated glycerol is die same backbone occurring naturally in, for example, animals and humans in mono-, di-, and triglycerides. Therefore, this compound might not necessarily be seen as foreign in the body.
The polymer preferably has an average molecular weight between about 1000 and 100,000 daltons, more preferably between 4000 and 40,000 daltons, depending, for example, on the molecular weight of the particular CSF-1 employed. Since the object of the modification is to obtain a conjugated protein witii retained biological activity, witii enhanced in vivo half- life over the unconjugated protein, and witii reduced immunogenicity, the molecular weight of the polymer will be chosen to optimize these conditions, e.g., a modified dimeric CSF-1 protein of over about 80 Kd apparent molecular weight. An additional advantage gained in derivatizing native dimeric
CSF-1 (i.e., non-recombinant protein) is tiiat the use of a scarce CSF-1 that is hard to purify would be maximized by such a modification.
Preferably the PEG homopolymer is substituted at one end witii an alkyl group, but it may also be unsubstituted. Preferably the alkyl group is a -C4 alkyl group, and most preferably a methyl group. Most preferably, the polymer is a monomethyl-substitutedPEG homopolymer and has a molecular weight of about 4000 to 40,000 daltons.
The covalent modification reaction may take place by any of the methods described above, preferably at about pH 5-9, more preferably 7-9 if 5 the reactive group on the protein is a free amino group. Using the latter approach, the protein is conjugated via at least one terminal reactive group added to die polymer. The polymer with die reactive group(s) is designated herein as "activated polymer". The reactive group(s) selectively react with a free amino or other reactive group of the protein. (If there is more than one
10 reactive group, d e conjugation conditions must be carefully controlled to prevent crosslinking; however, monovalent species are preferred.) The amount of intact activated polymer employed is generally about 1- to 30-fold molar excess of the active polymer over the protein. The exact molar ratio used is dependent upon a number of variables including die type of activated
15 polymer used, the pH, die protein concentration and die CSF-1 molecule used. For activated polymers oti er than the PEG-chloroformate active ester, d e molar excess of activated polymer over protein is preferably no more than about 11 moles per mole of CSF-1 dimer for derivatization of amino groups, and most preferably is about l to 8 moles per mole of CSF-1. The molar
20 excess of PEG-chloroformate active ester (PEG-PNP) over protein is preferably no more than about 5 moles per mole of CSF-1 dimer for derivatization of amino groups and most preferably is about 1 to 2 moles per mole of CSF-1.
Generally die process involves preparing an activated polymer
25 and thereafter reacting die protein with the activated polymer. Typically, the reaction is carried out in a buffer of pH about 7-9, frequendy at about 10 mM Hepes pH 7.5, 100 mM NaCl, if an internal-ester-free PEG-NHS reagent is used or at about 50 mM Na borate pH 9 if the PEG-PNP reagent is used, bodi of which are described below. The reaction is carried out generally at 0 to
30 25°C for from about 20 minutes to about 12 hours, preferably 25-35 minutes at 20°C or three hours at 4°C. Following the conjugation, the desired product is recovered and purified by column chromatography or the like.
The modification reaction with active PEG can be performed in many ways, described below, using one or more steps. Examples of suitable
35 modifying agents that can be used to produce the activated PEG in a one-step reaction include cyanuric acid chloride (2,4,6-trichlαro-S-triazine) and cyanuric acid fluoride.
In one preferred embodiment die modification reaction takes place in two steps wherein die PEG-OH is reacted first with an acid anhydride such as succinic or glutaric anhydride to form a carboxylic acid, and die carboxylic acid is tiien reacted with a compound capable of reacting with the carboxylic acid to form an activated PEG witii a reactive ester group that is capable of reacting witii the protein. Examples of such compounds include N- hydroxysuccinimide, sulfo-N-hydroxysuccinimide, 4-hydroxy-3-nitrobenzene sulfonic acid, and the like. Preferably, N-hydroxysuccinimide is used.
For example, monomethyl-substituted PEG may be reacted at elevated temperatures, preferably about 100-110°C for four hours, with glutaric anhydride. The monomethyl PEG-glutaric acid ti s produced is tiien reacted with N-hydroxysuccinimide in the presence of a carbodiimide reagent such as dicyclohexyl or diisopropyl carbodiimide to produce d e activated polymer, metiioxypolyethylene glycolyl-N-succinimidyl glutarate, which can tiien be reacted with the protein after purification. This method is described in detail in Abuchowski et al., Cancer Biochem. Biophvs..2:175-186 (1984). In another example, the monomethyl-substituted PEG may be reacted with glutaric anhydride followed by reaction with 4-hydroxy-3- nitrobenzene sulfonic acid (HNS A) in the presence of dicyclohexyl carbodiimide to produce d e activated polymer. HNSA is described in Bhatnagar et al., Peptides:Synthesis-Structure-Function. Proceedings of the Seventh American Peptide Symposium. Rich, et al. (eds.) (Pierce Chemical Co., Rockford IL, 1981), p. 97-100, in Nitecki et al., High-Technology
Route to Virus Vaccines (American Society for Microbiology: 1985), pages 43- 46 (based on talk November 8-10, 1984), entitled "Novel Agent for Coupling Synthetic Peptides to Carriers and Its Application", and in Aldwin et al., Anal. Biochem. (1987) 164:494-501. The disclosures of all of these are incorporated herein by reference.
As ester bonds are chemically and physiologically more reactive than amide bonds, it may be preferable to derivatize the protein with activated polyethylene glycol molecules that would not generate esters in the final product In one embodiment die PEG may be activated for attachment to the protein using PEG-amine or PEG-OH as starting materials. The PEG- OH may be converted to die PEG-amine as described by V.N.R. Pillar et al., J. Organic Chem.. 45_:5364-5370 (1980), the disclosure of which is incorporated herein by reference. Briefly, monomethyl PEG-amine (mPEG) is prepared by converting mPEG-OH to mPEG-tosylate and then to mPEG- phthalimide, and the phthalimide is cleaved with hydrazine to produce mPEG- NH2 in a Gabriel synthesis. The mPEG-amine is then reacted witii glutaric anhydride at room temperature for about four hours to produce mPEG- NHCO((2_2)3COOH. After the reaction the product is precipitated, purified, and reacted with N-hydroxysuccinimide and
dicyclohexylcarbodiimide to produce
Figure imgf000022_0001
This compound can then be reacted witii the appropriate free amino group(s) of the CSF-1 polypeptide.
In another embodiment, active ester forms of polyethylene glycol carboxylic acid useful for such conjugation are described in NitecM et al., Peptide Chemistry 1987 , Shiba & Sakakibara (Ed). Protein Research Foundation, Osaka (1988). Briefly, the active esters have a formula of:
R,-(0-CH2-CH2)n-0-(CHR2)-CO-OLG (I) or
R'-(0-CH2-CH2)n-l-0-CH2-CO-OLG (II) wherein R' is a lower alkyl group of 1-4 carbon atoms, R2 is H or an organic substituent n is about 8-500, LG is a leaving group selected from cyanomethyl, an aromatic group selected from a phenyl or naphthyl group substituted with from 1 to 5 substituents that render the aromatic group more labile, and a pyridyl group optionally containing 1-4 of these substituents. These esters may be produced, for Compound I, by alkylation of the polyethylene glycol with an alpha-haloalkanoic acid or ester thereof followed by esterification with HO-CH2-CN or the group corresponding to LG, or, for Compound (II), by oxidation of d e PEG to its acid, followed by esterification with HO-CH2-CN or the group corresponding to LG. Most preferably, Formula I is prepared and the activating agent is para-nitrophenol or ortho- nitrophenol. Most preferably, the polymer is conjugated to die protein via an amide linkage formed from die para-nitrophenyl ester of the polymer. For example, the PEG-OH may be converted to PEG-O- and reacted with BrCH2C02CH3, the methyl ester may be hydrolyzed, and die acid may be reacted witii j2-nitrophenol in the presence of dicyclohexyl-carbodiimide to produce PEG-0-CH2COO-^^NO2. This polymer is, following purification, in turn reacted with available free amino group(s) of the CSF-1. In another embodiment PEG-OH is reacted witii a chloroformate (also called a chlorocarbonate) to form a PEG active ester also called (PEG-PNP). After the PEG active ester is formed, it is reacted witii CSF-1 to form a PEG/CSF-1 conjugate. See also Veronese, gt aL. 1985, Biochem. and Biotech..11:141-152 which is hereby incorporated by reference in its entirety. Chloroformates may be purchased from companies such as Aldrich. They may also be made as shown in equation (1).
(i)
Cl- IC- CCll ++ RR--OOHH →→ CCll-C-O.-R + HC1
The chloroformate is made by reacting phosgene also known as carbonyl chloride with an alcohol (R-OH) which contains electron withdrawing substituents on the carbon that carries the -OH. The alcohol is preferably an acidic alcohol, more preferably an acidic alcohol which contains aromatic rings which have high extinction coefficients. Examples of R groups are: N-hydroxy-succinimide, N-hydroxy-sulfosuccinimides, cyanomethyl esters, all nitro, chloro, and cyano substitutions on benzene, naphthalene, or larger aromatic ring systems which may or may not contain hetero-atoms, such as pyridine, para-nitrophenol (PNP), ortho-nitrophenol (ONP), etc. Most preferred R groups are PNP and ONP.
After the chloroformate is formed, it is reacted with PEG-OH to form a PEG active ester as shown in equation (2).
9 9
PEG-OH + Cl-C-O-R → PEG-O-C-O-R + HC1
The chloroformate is reactive at two sites; at the bond between the chlorine and carbonyl group (more reactive) and die bond between the carbonyl and O-R group (less reactive). The more reactive site is where the chloroformate binds to the PEG. PEG-OH and the chloroformate are preferably added together at room temperature in an appropriate solvent, such as CHCI3, or CH2CI2. Preferably an acylation catalyst is added between 0 and 1 hours later, preferably the catalyst is pyridine or dimethyl pyridine. Preferably, the chloroformate is added up to 12 M excess, more preferably to a 2 M excess. The mixture is allowed to mix for preferably 4 hours, more preferably 16 hours. At this point, a precipitate may form. It is removed by filtration and discarded. Filtering devices such as Whatman glass fiber filters (GH/B) are acceptable. The resulting solution contains the PEG active ester as well as unreacted PEG and excess chloroformate. It is precipitated by adding an etiier, preferably the ether is dietiiyl ether. The precipitate contains the PEG active ester and can be washed witii appropriate solvents such as ether, redissolved and reprecipitated if necessary.
After the PEG active ester is formed, it is conjugated witii CSF-1 as shown in equation (3):
(3)
PEG i--OO--CC--OO--RR ++ NNHH2i--CCSSFF--1l→→ PPEEGG--OO--CC--1NH - CSF-1+ ROH
The chloroformate portion of the PEG active ester still has the less reactive site available. At this site, the covalent bond between the PEG and CSF-1 is formed. In the final product the PEG moiety is bound to CSF-1 by a
urethane, also called a carbamate, linkage ha ing the structure -0-C-NH-.
This linkage is relatively stable and will keep PEG conjugated to CSF-1 with little or no hydrolysis under physiological conditions.
Homodimeric recombinant CSF-1 from E. coli does not contain significant numbers of reactive free sulfhydryl groups after refolding has correctly proceeded to completion. The clone could be modified genetically, however, to include one or more novel cysteine residues that might retain sulfhydryl groups following refolding. The CSF-1 mutein so produced must still retain significant biological activity to be useful herein.
Altematively, free sulfhydryls can be generated by creation of selected heterodimers or by partial refolding of homodimers such that certain
SH groups, such as on cysi59, ~~e available for modification by activated polymers.
If the protein is being conjugated via a cysteine residue, a preferred mode of conjugation is as follows: mPEG-NH2 as described above is reacted at room temperature for preferably 0.5-1.5 hours with N-maleimido-
6-aminocaproic ester of 4-hydroxy-3-nitrobenzene sulfonic acid (mal-sac- HNSA), which is described by Nitecki et al., High-Technology Route to Virus Vaccines (Amer. Soc. for Microbiol., 1985), pp. 43-46, mentioned supra. The latter reaction is preferably conducted with about a 5-fold molar excess of mal-sac HNSA over PEG-NH2. After removal of hydrolysed or unreacted mal-sac HNSA (e.g., by dialysis, diafiltration, or size-exclusion chromatography), the reagent can then be reacted with the protein at room temperature in a buffer using equimolar amounts of reagent and protein. Other reagents, such as N-succmimidyl-4-(N-maleimidomedιyl)-cyclohexane-l- carboxylate (SMCC), or Xσ_2CO-NH(CH2)5-HNS A ester wherein X is Br, Cl, or I, can perform the same function as mal-sac HNSA under a variety of reaction conditions known to those skilled in die art.
Purification of Conjugates
After the conjugation reaction, a mixture of species, the identity of which will depend upon the reaction conditions, are likely present. These species which differ in d e number of PEG moieties conjugated to CSF- 1 may be separated by various methods including chromatography, electrophoresis and salt fractionation. Hydrophobic interaction chromatography (HIC) using phenyl-Sepharose has been shown to be particularly useful. Size separation may also be accomplished using non-reducing SDS-PAGE or molecular sieve chromatography. In addition, salting out of CSF-1 conjugates as well as conjugates of other proteins such as interleukin-2 (IL-2) has also been shown to be useful. Commonly used salts are ammonium sulfate, (NH4)2S04; sodium sulfate, Na24; magnesium salts and phosphates. The more highly conjugated species precipitated at lower salt concentrations than did less conjugated and unconjugated protein. Recycling ofpartially purified species through any of these techniques may result in a substantially homogeneous species of conjugated protein with regard to die number of polymers per molecule of protein.
Formulations The protein thus modified and optionally purified as to extent of conjugation is then formulated in a non-toxic, stable, pharmaceutically acceptable aqueous carrier medium, preferably at a pH of about 3 to 8, more preferably 5-8. Administration is by conventional protocols and regimens, preferably systemic, including intravenous administration. For in vitro applications, as for diagnostic purposes, the modes of administration and formulation are not critical. Aqueous formulations compatible with the culture or perfusion medium will generally be used. When used in vivo for therapy, the composition may include conventional physiologically acceptable excipients, such as water for injection, buffers, and stabilizers, as are known in the art A water-soluble carrier such as mannitol may optionally be added to die medium.
Modified CSF-1 may be used eitiier as the sole active ingredient or in combination with other proteins or compounds having complementary activity. Such other compounds may include antitumor agents such as adriamycin of lymphokines such as D -1, -2, -3, -4, interferons (alpha, beta or gamma), GM-CSF; G-CSF, and tumor necrosis factor. The effect of the modified CSF-1 may be improved by the presence of such additional components. A summary of formulation techniques for pharmaceutical compositions, including protein, is found, for example, in Remington's Pharmaceutical Sciences. Mack Publishing Co., Easton, PA, latest edition.
The dosage level of protein in the formulation will depend on die in vivo efficacy data obtained after preclinical testing and may vary depending upon die clinical application. The medium in which the protein is dissolved will be at a pharmaceutically acceptable pH when the mixture is reconstituted.
If the formulation is lyophilize _d, the lyophilized mixture may be reconstituted by injecting into the vial a conventional parenteral aqueous solvent such as, e.g., distilled water for injection.
The reconstituted formulation prepared as described above is suitable for parenteral administration to humans or other mammals in therapeutically effective amounts (i.e., amounts that eliminate or reduce the patient's pathological condition witiiout mortality or unacceptable morbidity) to provide tiierapy thereto. CSF-1 therapy may be appropriate for a variety of indications such as enhancing the immune system, enhancing cytotoxicity of macrophages, increasing monocytic and granulocytic white blood cell count treating infectious diseases such as cytomegaloviriis and bacterial infections (e.g., Gram-negative sepsis) by therapeutic or prophylactic administration to mammals, treating tumor burden such as sarcoma or melanoma in mammals, treating cholesterolemia (as described for GM-CSF by Nimer et al (1988) JAMA 260:3297-3300) and/or healing wounds in mammals. In addition, CSF-1 may be combined with G-CSF for stimulation of the immune system, as described in European Patent Publication No. 0273778 published July 6, 1988, d e disclosure of which is incorporated herein by reference. The dose and dosage regimen of the conjugated CSF- 1 will depend, for example, upon the phaπnacokinetics of the drug, the nature of the disease or condition, the type and length of polymer, the characteristics of the particular CSF-1, e.g., its therapeutic index, its spectrum of activities, the patient, and die patient's medical history. Different modified CSF-1 proteins are expected to have different pharmacokinetic and tiierapeutic properties that are advantageous for different routes of administration. A long-acting drug might only be administered every 3-4 days, every week, or once every two weeks. The clearance rate can be varied to give ultimate flexibility to fit the particular need of die patient by changing, e.g., the type of polymer, the size of the polymer attached, and the amino acid sequence to which the polymer is attached.
In the following examples, which illustrate the invention further, all parts and percentages are by weight unless otherwise noted, and all temperatures are in degrees Celsius.
Example I
Preparation of PEGylated CSF-1 Via Linkage Method One
A. Preparation of Activated PEG-NHS
A linear ester of monomethyl PEG of average molecular weight 7000 can be obtained by first reacting monomethyl PEG-7000, which is available from Union Carbide, widi glutaric anhydride at 100 to 110°C for four hours or by a method similar to that of Abuchowski et al.. Cancer Biochem. Biophvs..2:175-186 (1984), the disclosure of which is incorporated herein by reference. The resulting PEG-glutarate was reacted with N-hydroxysuccinimide in the presence of dicyclohexylcarbcdiimide, as described in detail by Abuchowski et al.. supra, on page 176. The resulting product is metiioxypolyethylene glycolyl N-succinimidyl glutarate, hereinafter designated as PEG*-7000. Reaction of PEG-4800 (available from Union Carbide) by the same metiiod resulted in PEG 800. A PEG-11,000 glutaramido NHS species (PEG"- 11,000) was prepared as follows, according to die procedure of Rajasekharam Pillai et al., J. Ore. Chem. 45_:5364-5370 (1980): A linear monomethyl PEG of average molecular weight 11,000 daltons obtained from Union Carbide (1.5 mmole) 5 was first dissolved in 10 ml methylene chloride and then 1.8 ml (22.2 mmole) of pyridine and 6.0 g (31.6 mmole) p-toluene-sulfonyl chloride were added. The flask was flushed witii nitrogen and the reaction mixture stirred at room temperature overnight The mixture was concentrated to about 5 ml and d e . product precipitated witii 75 ml ethyl ether. The precipitate was collected and
10 washed witii ether. The product (mPEG-tosylate) was recrystallized from ethanol.
The mPEG-tosylate (about 1.5 mmole) was dissolved in 20 ml dimethylformamide, and 2.5 g (17.0 mmole) potassium phthalimide was added. The solution was heated at reflux under nitrogen for four hours. The
15 precipitate that formed was filtered off and the filtrate was added drop wise to 300 ml ether to precipitate the product. The precipitate was filtered and washed witii ether. The product was suspended in 30 ml methylene chloride and stirred for 0.5 hours. Insoluble impurities were filtered off and die product (mPEG-phthaiimide) was precipitated with ether. Next the mPEG-
20 phthalimide (about 1.1 mmole) was dissolved in 15 ml ethanol and 2.0 ml (41.2 mmole) hydrazine hydrate was added. The mixture was refluxed overnight The reaction mixture was cooled to room temperature and die product was precipitated witii ether.
The precipitate was collected by filtration and resuspended in
25 25 ml methylene chloride. Insoluble impurities were filtered off and the product was precipitated with etiier. This precipitate, mPEG-1 l,(XX)-amine, was suspended in CH2CI2, filtered, and precipitated with etiier two more times. The second time it was completely soluble in methylene chloride.
A total of 0.5 g of the mPEG-ll,000-amine was dissolved in
30 10 ml dioxane to which was added 0.25 g glutaric anhydride. The reaction was carried out for four hours at room temperature. After the reaction, the product, mPEG-NHCO(CH2)3COOH, was precipitated with about 100 ml ether. The mixture was filtered, and d e product was redissolved in CH2CI2, filtered into ether, filtered and dried. The yield of product was 200 mg.
35 The mPEG-NHCO(CH2)3COOH was men reacted with N- hydroxysuccinimide in the presence of dicyclohexylcarbodiimide, as described above for preparing PEG*-7000. The resulting product is designated herein as PEG"- 11,000.
B. Purification and Refoldin of CSF- 1
An E. coti strain HW22, transformed with the plasmid pJN653 containing the SCSF/NV3CV158 gene (the plasmid being deposited as ATCC
No. 67,561 on November 12, 1987) was grown in a 10-liter fermenter in basal medium containing 96 mM (NF_4)2S04, 28 mM KH2PO4, 4 mM Na3 citrate.2H20, 1.7 ml l TK9 (30 mM ZnS04, 30 mM MgS04, 1 mM CuS04), with sterile additions of 6.5 g 1 glucose, 2.2 mM MgSθ4.7H2θ, 95 μM, FeSθ4.7H20, and 26 mg 1 thiamine.HCl at 30°C until an OD680nm of 10 was reached. Casamino acids were tiien added to 2% w/v. CSF-1 expression was induced by shifting the temperature of the culture to 37°C. After four hours the absorbance at 680 nm reached 79.
The cells were harvested by 5-fold concentration and diafiltered against ten volumes of 5 mM EDTA, pH 8.5, using Dorr-Oliver tangential cross-flow microporous filtration. The cells were disrupted by ti ree passes at 7,500 psi in a Manton-Gaulin high pressure mechanical cell homogenizer. 1- Octanol was added to 0.1% (v/v) and d e homogenate was held overnight at 4°C. The homogenate was made 25% sucrose by addition of a 63% w/v sucrose solution. The insoluble protein fraction (refractile bodies) was separated from cell debris by continuous flow disk stack centrifugation (Westphalia SB7) at 9000 xg, 1 liter/minute and 4-6°C. The wet pellet was mixed 50:50 (w/v) with deionized water and stored at -20°C in aliquots. Twenty-five grams of refractile body suspension
(approximately 390 mg of protein) were solubilized in 250 ml of 8 M urea containing 25 mM Tris, 10 mM sodium phosphate buffer (pH 8.4), 1 mM ethylenediamine tetraacetic acid (EDTA), and 4 mM did iothreitol (DTT). After two hours at room temperature, the solution was clarified by centrifugation at 15,000 x g for 15 minutes. A 150-mI aliquot of the solubilized CSF-1 was tiien loaded onto a 5 x 8 cm DEAE-Sepharose (Pharmacia) column equilibrated in 6 M urea containing 25 mM Tris, 10 mM sodium phosphate buffer (pH 7.0). The column was washed with 1 bed volume of die above solution, which had been modified to contain 1 mM DTT and 1 mM EDTA, and the CSF-1 was then eluted with a 1.4-liter salt gradient of 0-0.6 M sodium chloride in the wash buffer. The CSF-1 peak eluted at approximately 0.06 M sodium chloride.
The remaining 90 ml of solubilized refractile bodies was tiien purified over die DEAE-Sepharose column in identical fashion. The combined CSF-1 pools (165 ml) contained approximately 250 mg of protein at a purity of approximately 50%.
The CSF-1 was then refolded by diluting die DEAE-pool into refolding buffer containing 50 mM Tris (pH 8.5), 5 mM EDTA, 2 mM reduced glutathione, 1 mM oxidized glutathione, pre-cooled to 4°C. The CSF- 1 was allowed to refold for 30 hours at 4°C. The pH of the refolded CSF-1 was tiien adjusted to 6.8 using 8.5% phosphoric acid solution. The solution was then clarified by centrifugation for 10 minutes at 15,000 x g and loaded onto a 5 x 4 cm DEAE Sepharose column pre-equilibrated in 10 mM sodium phosphate, 25 mM Tris (pH 6.8). The column was washed with 300 ml of this buffer and tiien eluted witii a 700 ml, 0-0.6 M sodium chloride gradient in die same buffer system. The CSF-1 eluted at approximately 120 mM sodium chloride. Ammonium sulfate (4 M stock, pH 7.0) was then added to the 95- ml DEAE pool to a final concentration of 1 M. The CSF-1 was then filtered through a Nalgene 0.45 micron filter and loaded (at 4°C) onto a 21.5 x 150 mm Bio-Rad TSK Phenyl-5-PW column equilibrated in depyrogenated 1.5 M ammonium sulfate, 0.1 M sodium phosphate (pH 7.0). The column was washed witii two bed volumes of this loading buffer and tiien eluted in 0.1 M sodium phosphate (pH 7.0) using a 45-minute gradient in which the ammonium sulfate concentration decreased from 1.5 M to 0 M and die etiiylene glycol concentration increased from 0-60% (v/v). All operations were carried out at 4°C under essentially pyrogen-free conditions. The CSF-1 eluted at approximately 0.6 M ammonium sulfate in 30% ethylene glycol. The CSF-1 was then extensively dialyzed into 10 mM Hepes buffer (pH 7.5) containing 150 mM sodium chloride and subsequendy was filter sterilized through a Millex 0.45 micron filter.
Approximately 50 mg of purified SCSF/NV3CV158 CSF-1 was obtained. Greater tiian 90% of die final CSF-1 product migrated as a single species on SDS-PAGE, and die same product was approximately 96% one species as analyzed by RP-HPLC in acetonitrile/TFA. The specific activity was about 1.5-1.7 x 108 units/mg (units determined as colony fomiing units equivalents using a CSF-1-dependent cell line, and protein concentration determined using A28O nm and an extinction coefficient of 0.6, estimated from amino acid composition determinations). This specific activity is at least equivalent to, if not greater than, that of purified native Mia PaCa CSF-1. The endotoxin content of die refolded, purified CSF-1 product, determined by LAL assay, was 0.5-1 ng/mg of CSF-1.
In a similar manner, IL ____li protein produced under control of the PL promoter from DNA encoding SCSF/ V2CV150 as described in
European Patent Publication No. 0272779 was refolded and purified in a similar manner. The vector used to transform the E. coli was prepared by substituting the appropriate synthetic fragment for the excised HmdHI/BstXI
DNA of the appropriate CSF-17 vector to encode the CSF/NV2, lacking N- terminal glu-glu.
C. Conjugation of PEG*-7000 to CSF-1
The CSF-1 protein (SCSF/NV2C 150) in Section B was dialyzed (1 mg protein/ml) into a medium of 10 mM Hepes buffer, pH 7.2 containing 100 mM NaCl. The PEG*-7000 was dissolved rapidly in a small volume of water and immediately added to the protein solution, which was continuously stirred throughout the reaction. A 3-fold to 30-fold molar excess of PEG*-7000 over CSF-1 dimer was employed. Reaction was complete in about 30 minutes at 20°C and in about three hours at 4°C.
The samples were analyzed by size exclusion HPLC. Figure
3A shows the chromatogram of underivatized rCSF-1 (SCSF/NV2CV150) sample prior to the experiment. Figure 3B shows the chromatogram of 100 μg of the same CSF-1 sample after derivatization with a 5-fold molar excess ofPEG*-7000 for 30 minutes at 20°C. Radioimmunoassay, performed according to die method of Stanley, R. and Guilbert, J. Imm. Methods.
42:253-284 (1981), confirmed tiiat the first three major absorbance peaks observed in Figure 3B contained rCSF-1. See Table I.
The fractions 30, 32, 34, 36, 37, 38, 39, 41 and 42 were also analyzed for biological activity, using the mouse bone marrow assay described by Moore et al., J. Immunol. (1983) 131:2397 and by Prystowsky et al., Am.
J. Pathol. (1984) 114:149. the disclosures of which references are incorporated herein by reference. Table I provides the results. TABLE I
Derivatization with PEG*-7000 Clone: SCSF/NV2CV150
Figure imgf000032_0001
5 ^Fractions were assayed after dilution to about 1000 RIA U/ml. lUnits/ml
2Bioactivity ratio = bioassay divided by RIA ND = Not -determined
The results in Table I and those below show that the unreacted o rCSF-1 and die smallest molecular weight peak of derivatized rCSF-1 retained approximately full bioactivity (within experimental error). Larger species retained much less residual bioactivity. The reaction with PEG*-7000 apparendy did not significantly affect the radioimmunoassay (RIA) reactivity of CSF-1, because the unfractionated, derivatized rCSF-1 retained 5 approximately full RIA immunoreactivity per mg of protein.
D. Conjugation of PEG 1.000 to CSF-1
Biologically active protein was purified and refolded after expression of a construct encoding SCSF/CV150 in E. coli under control of the P promoter in a vector constructed as described in European Patent Publication No. 0272779 published June 29, 1988. The strain used was an R coli λ lysogen, DG116, transformed with the plasmid O/E pPi, CSF- 17 asp59/CV150 (ATCC No. 67,389). The protein was produced intracellularly in a monomeric, insoluble form, and purified and refolded as described in PCT Publication No. WO 88/08003 supra.
A total of 2 mg of this purified CSF-1 at 1 mg ml protein was dialyzed into Hepes buffer at pH 7.2 containing 100 mM NaCl. The PEG"- 11,000 was dissolved rapidly in a small volume of water and immediately added to die protein solution, which was continuously stirred diroughout the reaction. An 8-fold molar excess of PEG"- 11,000 over CSF-1 dimer was employed. Reaction was completed in 30 minutes at 20° C. This CSF-1 reacted witii the PEG"-11,000 in a fashion very similar to the reaction of
SCSF/NV2CV150 with the PEG*-7000. As Table π shows, mild PEGylation (1-2 per dimer) again had minimal effect upon bioactivity, while the highly modified pool had significandy reduced activity.
TABLE H
Comparison of Derivatization of rCSF-1 With PEG"- 11,000 and PEG*-7000
Figure imgf000033_0001
lEstimated from apparent native molecular weight as measured by SEC-HPLC and SDS-PAGE.
Figure 4 shows a size exclusion HPLC chromatogram of 8 mg of rCSF- 1 (SCSF/CV150) derivatized with PEG"-11,000. The mildly derivatized fraction, pooled as shown in the figure, was found to retain essentially full biological activity, and to migrate with an apparent molecular weight of 80,000 daltons on sizing, and 45,000 daltons on nonreduced SDS-PAGE (Figure 8). This fraction was recovered at an overall yield of about 20%.
The difference in sizes of PEG-CSF estimated by these two techniques is consistent with observations made by od ers. PEG can alter the ability of a protein to bind SDS, affecting mobility on SDS-PAGE; and it can also affect size exclusion estimations, e.g., by hydrophobic interaction with d e column matrix.
The endotoxin level of this sample, as assayed by d e Limulus amebocyte lysate (LAL) assay, was found to be less than 1 ng/absorbance unit at 280 nm (A28O unit) protein. (LAL assay is described by a product brochure (1982) for PyroteE brand of LAL available from Associates of Cape Cod, Inc., Woods Hole, MA; it is also described by Watson et al. (eds.) "Endotoxins and Their Detection with the Limulus Amebocyte Lysate Test", Proceedings of an International Conference on Endotoxin Standards and Limulus Amebocyte Lysate Use with Parenteral Drugs. Alan R. Liss, Inc., New York (1982); and by Levin et al. (1964) Bull. Johns. Hopkins Hosp.. 115:265.)
E. Conjugating of PEG-.-4800 to CSF-1
The same conditions used for PEG"-11,000 were used to react the rCSF-1 from the clone SCSF/CV150 with PEG*-4800. The CSF-1 was successfully derivatized, and die pool that was mildly derivatized (at one or two sites) retained essentially full bioactivity.
F. Pharmacokinetics of PEGylated CSF-1 and Unmodified CSF-1 in Rats
Three male CD rats (Charles River Breeding Labs, Wilmington, MA) of average weight 161 g were injected in the tail vein with 1 mg/kg of the mildly derivatized fraction of PEG"-11,000-derivatized CSF-1 from the SCSF/CV150 clone described above and shown in Figure 8. Blood plasma samples were collected by an indwelling cadieter, and CSF-1 titer was determined by RIA. Urine samples were collected at 0, 30, and 120 minutes and assayed. Additional data were also collected using the underivatized rCSF- 1 andrCSF-1 expressed in mammalian cells (COS) that arises from a glycosylated 522-amino acid precursor (LCSF). The rats in the experiments with unmodified CSF-1 had an average weight of 178 g and were injected with 125 μg/kg of the unmodified CSF-1. All other conditions were the same.
Figure 5 compares the time courses of blood clearance of modified and unmodified CSF-1 protein. The systemic clearance is calculated by dividing die dose by die area under die blood plasma curve. The data show tiiat the systemic clearance of derivatized rCSF-1 in d e blood is 0.302 ml/min kg versus 3.84 ml/min/kg for underivatized rCSF-1. This represents a 12.7-fold extended residence time of die derivatized as compared to die underivatized protein. Figure 6 shows size exclusion HPLC of the rat blood plasma
120 minutes after injection of the derivatized CSF-1 (SCSF/CV150). This figure shows that the RIA-detectable rCSF-1 signal in the blood plasma was 43-80 Kd in apparent size two hours after intravenous injection. This observation suggests that the RIA signal that was measured in d e pharmacokinetic experiment (at 120 min.) represents intact PEG-rCSF- 1 and rCSF-1. Westem blotting by the technique of Bumette, (1981) Anal. Biochem, 112:195-203 (developed with an antiserum to recombinant CSF-1 capable of detecting CSF-1 fragments, followed by '-~I-pτotein A) of a non- reduced SDS-PAGE gel of urine and plasma verified tiiat the rCSF-1 antigen detected by Westem blotting was apparently intact, dimeric, and die same size as the material that was injected.
Example TI
Preparation of Activated PEGylated CSF-1 Via A Second Linkage Method A. Preparation of PEG-Ester
1. The carboxymethyl derivative of mPEG-5000 was prepared: '
Sodium-naphthalene was prepared by addition of 0.15 g Na to a solution of 0.64 g naphthalene in about 20 ml tetrahydrofuran (THF) freshly distilled from sodium benzophenone. Monometi ylpoly(ethylene glycol) of average molecular weight of 5000 ("mPEG 5000") was dried overnight in a vacuum dessicator with P2O5. The Na-naptiialene solution was added dropwise to a solution of 2.5 g mPEG 5000 in -50 ml dry THF (freshly distilled). When the green color persisted in the solution to indicate excess, base addition was ceased and 1.2 ml BrCH2COOCH3 was added dropwise. The green disappeared and die mixture became cloudy. The mixture was stirred overnight at room temperature. The cloudy mixture was poured into a flask containing about 70 ml cold etiier. The precipitate was collected by vacuum filtration and washed witii ether. The dry solid was dissolved in 75 5 ml 1 M NaOH and stirred at room temperature for 2.5 hours to hydrolyze the methyl ester. The pH was adjusted to about 3 by addition of HC1 and d e solution concentrated on a rotary evaporator. The residue was taken up in CH2CI2 and stirred for about one hour. Insoluble material was filtered off and die solution was poured into etiier. The solid was collected by vacuum
10 filtration, washed with ether, and dried in a vacuum dessicator over P2O5. This yielded d e desired carboxymethyl mPEG-5000 acid. The acid was titrated to demonstrate tiiat complete conversion had taken place. The preparation of carboxymethyl mPEG-5000 was repeated at a larger scale. The precipitate was collected and dried. Yield 8.5 g. Titration showed about
15 102% acid. The reference for this experiment is Buckmann et al., Makromol. Chem. 182:1379 (1981).
2. Para-nitrophenyl ester of carboxymethyl mPEG-5000 was prepared:
A total of 1 g mPEG-5000 acid (2 x 10-4 moles) was dissolved in 3 ml CHCI3. To this solution was added 0.28 g β-nitrophenol (2 x 10-4 20 moles). The solution became pale yellow. Then 0.041 g of dicyclohexyl carbodiimide (DCC) (2 x lC moles) was dissolved in a small amount of
CHCI3 and added dropwise to die PEG-acid solution at room temperature.
After about 10 minutes of stirring, 2 μl of the CHCI3 mixture was added to
1.0 ml 0.01 M phosphate buffer, pH 7.0. The absorbance at 400 nm of the μ- 25 nitrophenol anion was 0.2443. 5N NaOH was added, increasing the A400 to
0.5847 (% ester is 58.2% as calculated by die formula below:
%
Figure imgf000036_0001
After about three hours of reaction 1 μl of the CHCI3 mixture
30 was added to 1.0 ml of 0.01 M phosphate, pH 7.0. The A400 was 0.2238.
When 50 μl 5N NaOH was added, the A400 was 1.154, yielding 80.6% ester.
A precipitate, dicyclohexylurea, appeared and was filtered off through a glass fiber filter and washed witii CHCI3. The ester was
35 precipitated by adding about 300 ml anhydrous etiiyl ether. The mixture was allowed to precipitate for about three hours and was tiien filtered tiirough a glass frit. The precipitate was then redissolved in CHCI3, reprecipitated witii about 100 ml ethyl ether, and filtered through a medium glass frit. A small amount of damp solid was dissolved in 0.01 M phosphate buffer, pH 7.0. The A400 was 0.0240; when 50 μl 5N NaOH was added, d e A400 increased to 3.330 (% ester was 99.3).
The main precipitate was dried in a vacuum desiccator overnight The flask in which it was contained was washed witii water and the residues were lyophilized. A total of 4 mg of the precipitate was then dissolved in 2 ml 0.01 M phosphate at pH 7.0 (A400 = 0.0276). When 50 μl 5N NaOH was added, the A400 was 3.50 (off-scale). A total of 200 μl of the solution was diluted to 800 μl 0.01 M phosphate, pH 7.0. The A400 was 0.7985. Calculated % ester = 99.3.
A total of 1.5 mg of lyophilized residues was dissolved in 1.0 ml of 0.01 M phosphate, pH 7.0. The absorbance was 0.0680. A total of 50 μl of 5N NaOH was added and the A400 was 1.106 (% ester - 93.9). The residues on the filter from the main precipitate were washed witii water and lyophilized over a weekend. The weight was 131 mg of fluffy white powder. A small amount was dissolved in 0.01 M phosphate, pH 7, and die A400 was 0.0859. On adding 50 μl 5N NaOH, the A400 was 0.6794 (87.4% ester).
Structure of ester:
PEG- -OO--CCHH22 $-C-oC<o} )*N«02 feNP)
B. PEGylation of CSF-1 faspsoSCSF/CV150)
The para-nitrophenyl ester just described was coupled with die asp59SCSF/C 150 refolded protein described in Example I as follows: The rCSF-1 (200 μg at 1 mg ml) was dialyzed into 20 mM
Hepes buffer, pH 7.2, containing 100 mM NaCl. A total of 0.8 mg of the para-nitrophenyl ester was dissolved in a small volume of water and 259 μg of this solution was immediately added to the SCSF/C 150. The conjugation was carried out at 20°C for four hours, and the samples were analyzed by size exclusion HPLC. The mildly derivatized rCSF-1 (corresponding to about 1 or 2 PEG molecules per CSF-1) retained essentially full bioactivity as assayed on mouse bone marrow.
If the reaction is carried out in cuvettes in a dual-beam Hewlett- Packard spectrophotometer, release of para-nitrophenol anion can be monitored, permitting the coupling reaction to be stopped reproducibly after a given amount of release has occurred.
Example iπ
Preparation of PEGylated CSF-1 Via a Third Linkage Method
A. Preparation of Activated PEG-Ester (PEG-PNP)
25 grams (2.5 mmole) of monomethyl PEG of average molecular weight of 10000 (m-PEG 10,000; Union Carbide) containing only low concentrations of tiiol was dissolved in 250 ml CH2CI2 in a 500 ml 3 neck round bottom flask. 5 grams (25 mmoles) p-nitrophenyl chloroformate (PNP-chloroformate) and 3 grams (25 mmole) dimetiiylaminopyridine (DMAP) were added. The mixture was stirred overnight under nitrogen at room temperature. The reaction mixture was filtered to remove DMAPHCI and concentrated to 100 ml. The concentrated solution was added to 1 liter etiier with stirring. The precipitate was collected on a coarse scintered glass filter. The precipitate was then stirred with about 400 ml methylene chloride for approximately 1 hour and tiien filtered and concentrated to about 100 ml. The PEG ester was precipitated again by addition of die solution to 1 liter ether. The precipitate was collected using a glass fiber filter and dried in a vacuum dessicator. The yield was 17.5 grams. The product was assayed for p-nitrophenyl carbonate content
10.3 mg (1.03 x 10-6 mole) was dissolved in 10 ml 0.1 M sodium phosphate, pH8. Free or unreacted p-nitrophenyl (PNP) impurity was determined by absorbance at 400 nm. The initial A400 was 0.37. Addition of 50 microliters of 5N NaOH to 1.0 ml PEG-PNP hydrolyzed all ester present and A400 was 2.06. If all the product weighed (10.3mg) had been PEG-PNP, (i.e. no free PNP was present) 1.03 x lO-4 mole/liter PNP would have been released having an A400 = 1.85 which is referred to as die tiieoretical maximum A400. Therefore 2.06 -0.37 x 100 = 91% of the material weighed was actually PEG- 1.85
PNP. Since the initial absorbance (0.37) was rather high, free PNP was apparendy present. The product was further purified by dissolving in about 75 ml CH2CI2 and reprecipitating by addition to 1.2 liters ether. The precipitate was collected, dried (15.8 grams) and reassayed.
A400 after NaOH - A400 before NaOH
__ x l00 = theoretical maximum A400 1.78 - 0.157
. x 100 = 89%
1.82
B. Preparation of PEGylated CSF-1 CSCSF/NVCV158)
CSF-1 was produced essentially as described in Example I. CSF-1 was concentrated to about 10 mg ml in 0.05 M Na borate, pH 9.0. 5 mg PEG-PNP obtained as a solid from Example HI Part A was added per ml of CSF-1. (approximately a 1:1 M ratio). The reaction was continued for 2 hrs at room temperature.
NaCl was added to the reaction mixture to a final concentration of about 5M (saturation) and the high salt mixture was loaded onto a phenyl Sepharose (Pharmacia Fast Flow High Substitution) column. Approximately 1 ml of phenyl Sepharose was appropriate for 10 mg protein loaded. CSF-1 was eluted from die column with an ammonium sulfate gradient from 1.2 - 0 M in 0.1 M Tris, pH 8.5. Fractions were analyzed by Coomassie-stained non-reducing SDS-PAGE and bioactivity in the NFS 60 bioassay was determined. The results are shown in Table HI. The pluses show relative amounts of different CSF-1 conjugates in the samples. The bioactivity data is die average of three assays. TABLE m
Figure imgf000040_0001
Example IV
A. Preparation of PEG ylated CSF-1 (SCSF/NV3CV158)
Growth and harvest of E* coli strain HW 22 transformed witii pJN653 containing the SCSF/NV3CV158 gene was as described in Example I.
The refractile body suspension was then solubilized, refolded, and purified also as described in Example I. The final specific activity was about 1.5 x 10s units/mg in a CSF-1 bioassay performed on a CSF-1 dependent cell line.
At a concentration of 2.6 mg ml in 10 mM Hepes buffer, pH 7.5 and 100 mM NaCl, 40 mg of the refolded CSF-1 above was incubated with stirring at 20°C. PEG"-11,000 was dissolved in 200 μl of distilled water and added immediately to the CSF-1 solution at an 11-fold molar excess of PEG"-11,000 over CSF-1 dimer (the PEG"-11,000 was approximately 55% non-hydrolyzed and active). After 30 minutes of incubation, the reaction was stopped by addition of 2 moles of ε-aminocaproate per mole of PEG"- 11,000, from a 1 M stock solution. The sample was concentrated to 6 ml by Amicon Centricon-30 centrifugation and purified by size exclusion HPLC in tiiree identical 2-ml-load runs. The column used was Bio-Sil® TSK-250, 600 x 21.5 mm (BioRad), equilibrated in 0.2 M Na2HPθ4/NaH2P0 buffer pH 7.0. The A28O peaks of mildly PEGylated protein in the three runs were pooled (pool 1, SEC) and concentrated to a final 2-ml volume, which was reinjected on the same column. The active, PEGylated fractions were pooled and concentrated.
The final pool (pool 2, SEC), representing 15% of the starting material, consisted of 6 mg of CSF-1 that had approximately 100% of die initial bioactivity of d e unmodified CSF-1 and contained a major species of PEG-CSF-1 that migrated at about 45 K apparent Mr on SDS-PAGE. Small amounts (about 10%) of unmodified CSF-1 and more highly modified CSF-1 remained in the pooled product Characterization of size exclusion chromatography (SEC) pools 1 and 2 is shown in Tables IV and V tables below:
Figure imgf000041_0001
TABLE V
Bioassay (using Bioassay (using RIA (as described a CSF-1 dependent a CSF- ldependent above) ("units"/ cell line) (units/ cell line)(units/ Sample Aϊgp unit A^ unit) mg
Unmodified rCSF-1 1 x 108 1 x 108 1.5 x 108
Pool 2, SEC 2.7 x 108 1.1 x 108 1.6 x 108 B. Pharmacokinetics of PEGylated CSF-1 (SCSF NV3CV158)
The same conditions as described in Example I were used to estimate the average intravenous clearance rate in three rats of CSF-1 and PEG- CSF-1 described in Section A of this example, except that the doses for both PEGylated and non-PEGylated CSF-1 were 6 mg/kg.
Figure 7 shows the curve of relative concentration of CSF-1 from the blood of three rats versus time after intravenous injection. The clearance was found to be about 0.63 ml/min/kg for PEGylated CSF-1 and 7.50 mVmin/kg for non-PEGylated CSF-1 (tiiree hours as opposed to five minutes). This represents about a 12-fold increase in average residence time in the blood for die PEGylated molecule.
Example V Ammonium Sulfate Fractionation of PEG-CSF-1 Conjugates A. Small Scale 10 ml of reaction mixture containing OPEG-CSF-1
.(unconjugated CSF-1), 1 PEG-CSF-1 (1 mole PEG/mole CSF-1), 2PEG- CSF-1 (2 moles PEG/mole CSF-1), 3PEG-CSF-1 (3 moles PEG/mole CSF-1) and 4PEG-CSF-1 (4 moles PEG/mole CSF-1) was approximately 1 mg/ml protein. Solid (NILt^S φ was added to about 1.3 M. A precipitate formed and was removed by centrifugation at 10,000 rpm for 10 min. The pellet was saved and (NU )2S04 was added to the supernatant until it became turbid at about 1.4M. The precipitate was removed by centrifugation. This procedure was continued making additional (NH4)2S04 cuts at about 1.5, 1.6 and 1.7M. Precipitates were resuspended in 0.1M Tris pH 8.6. SDS-PAGE analysis of the precipitates showed enrichment of unconjugated CSF-1 in the 1.7 M( H )2S04 precipitate and of die lPEG-CSF-1 species in the 1.5 and 1.6 M precipitates. The 2PEG-CSF-1 was the predominant species in the 1.4 M precipitates although 1PEG- and 3PEG-CSF-1 were also present Table VI shows the relative amounts of various CSF-1 conjugates in pellets of increasing (NH4)24 concentration as analyzed by Coomassie stained non¬ reducing SDS-PAGE. Repeated (N__4)24 fractionation of fractions containing more than one species of CSF-1 conjugate resulted in essentially pure unconjugated CSF-1 and lPEG-CSF-1. This repetition can also be used to produce substantially pure 2PEG-CSF-1 or 3PEG-CSF-1. TABLE VI Ammonium Sulfate Fractionation of PEG-CSF-1 Conjugates
Fraction CSF-1 Conjugate Present (#PEGs/CSF-l Dimer)
0 1 2 3 4 1.3M + + + + +
1.4M + + ++ +
1.5M + ++++
1.6M + ++++
1.7M +++++
B. Large Scale
Approximately lg of CSF-1 (SCSF NV3CV158) was conjugated according to Example HI. After fractionation by (NH)2S04 as described in Part A, approximately 600 mg of CSF-1 having the molecular weight of CSF-1 dimer conjugated to one PEG was purified. This conjugate had a specific activity of about 6.2 X 107 U/mg. About 80 mg of a mixture of CSF-1 conjugates having 1, 2 or 3 PEGs linked was also recovered and had a specific activity of about 3.2 X 107 U/mg.
Example VI
Ammonium Sulfate Fractionation of PEG-IL-2 Conjugates A. PEG-IL-2 Conjugates having an Amide Linker
Approximately 12 mg of IL-2 which was produced and purified essentially as described in PCT Patent Publication No. WO 88/08849 published November 17, 1988 and PEGylated essentially as described in U.S. Patent No. 7,766,106 was resuspended in 1 ml 1.0M Tris pH8. Solid (NH)2S04 was added until die solution became turbid. The sample was centrifuged at 12,000 rpm for 12 min. The pellet was resuspended in 0.1 M Tris pH 8.6. (NH)2S04 fractionation was continued as described in Example V and SDS-PAGE analysis of resuspended pellets indicated tiiat unconjugated EL-2 could be separated from conjugated IL-2. Furthermore, similar to the results described in Example V for CSF-1, fractions were obtained tiiat were obtained tiiat were enriched for IL-2 conjugated to 1PEG. Recycling of fractions enriched for a particular conjugate is expected to result in substantially pure conjugate. B. PEG-IL-2 Conjugates having the Urethane Linker
PEG-IL-2 was obtained by d e procedures described in commonly owned copending U.S. Serial No. filed on January
20, 1989. The PEG moiety was attached to IL-2 via the urethane linker herein described. (NH)2S04 precipitation essentially as described in Example VI Part A and in Example V resulted in fractionation of the various PEG-IL-2 conjugates.
Example YTI
In yjyo Efficacy of PEGylated CSF-1 (SCSF NV3CV158): bacterial 0 infection model
Groups of five mice were injected intraperitoneally (ip) on day -
1 (a day prior to infection with a letiial dose of £. coli SM18) with die
SCSF/NV3CV158 described in Example HI, PEGylated SCSF/NV3CV158 prepared as described in Example HI, or saline (control group). Two CSF-1 5 dosage groups (10 μg/mouse and 50 μg/mouse) were used. At day 0, all mice were injected ip witii a letiial dose (5 x 10? cells) of R ecu SMI 8. The number of mice surviving was followed for five days. The results are shown in the table below:
TABLE VH
- Number of Mice Surviving after day:
1 2 3 4 5 6
Control (saline injected) 2 0 0 0 0 0 Unmodified CSF-1
10 μg 3 2 2 2 2 2 5 50 μg 3 3 3 3 3 3
PEGylated CSF-1 10 μg 2 2 1 1 1 1
50 μg 5 4 3 3 3 3
The results show that there is a dose-dependent effect of rCSF- 0 I on survival. The slight difference between the modified and unmodified CSF-1 in efficacy in this single experiment is not statistically significant PEGylation of CSF-1 did not reduce die efficacy detected by this protocol, nor did it increase drug-related toxicity observable in this experiment
Example VIII
In Vivo Test of Conjugated CSF-1 for Anti-Tumor Efficacy
A. Meth A Sarcoma Modgl
PEGylated CSF-1 is injected intraperitoneally, subcutaneously or intravenously (i.p., s.c, i.v.) at up to 50 μg/dose per mouse (10 mice per group) implanted subcutaneously with a Meth A sarcoma tumor 7 days earlier. Dosing schedules depend upon the particular PEGylated CSF-1 preparation used. Tumor volume is followed for 14 days after the beginning of CSF-1 treatment. The results are evaluated by comparing the mean change in tumor volume* over time in the CSF-1 treated and control treated mice. *(ΔTV =
Ratio of the mean tumor volume at the day indicated to die mean tumor volume at day 0 within a single group of mice).
B. B16 Metastases Model
A second in vivo tumor model using PEGylated CSF-1 to prevent metastases in the B 16 murine melanoma cell line is also useful.
1-10 x 104 tumor cells, suspended in 0.2 ml of Ca+2 and Mg+2- free HBSS, are inoculated into unanesti etized mice in the lateral tail vein. From 14 to 21 days after tumor cell inoculation, the mice are sacrificed and necropsied. During necropsy, the lungs and brain are removed, rinsed in water, and weighed. The lungs are then fixed in Bouin's solution, and die number of surface tumor nodules per pair of lungs are determined witii the aid of a dissecting microscope.
Recombinant human CSF-1 PEGylated prepared according to Example HI and having a potency of 1-25 x 107 U/mg and pharmacologically acceptable endotoxin levels is used. CSF-1 is freshly obtained prior to each experiment from frozen stocks and diluted immediately prior to injection in USP 0.9% saline. PEGylated CSF-1 is delivered i.v., i.p. or s.c. on schedules which are dependent upon d e particular PEGylated CSF- 1 CSF-1 preparation used. The dosing levels used range up to 5mg/kg. As a negative control consisting of a non-specific and non-tiierapeutic protein, either USP human serum albumin (HSA) or boiled PEGylated CSF-1 is used. PEGylated CSF-1 is boiled for 30 min to inactivate the CSF-1 activity. The efficacy data demonstrates that PEGylated CSF-1 produces a significant reduction in die median number of pulmonary metastases.
Example IX
In Vivo Treatment of CMV Infection with PEGylated CSF-1 Outbred CD- 1 mice are treated i.p., i.v. or s.c.with PEGylated
CSF-1 at doses of up to 400 μg/kg, starting two days before infection with a sub-lethal dose of cytomegalovirus (CMV). Dosing schedules are dependent upon the particular PEGylated CSF-1 preparation used. Mice are sacrificed on the tiiird day after infection and d e extent of viral replication in target organs such as the spleen is evaluated by plaque assay. The results show that mice treated with PEGylated CSF-1 have significantly lowered organ viral titer compared to the saline-treated control mice, indicating that CMV infection is less severe in PEGylated CSF-1 treated mice.
Separately, PEGylated CSF-1 may be tested in a lethal murine CMV infection model in outbred CD- 1 mice (this is in contrast to the above experiment using sub-lethal doses of CMV, in which organ titers are monitored). When PEGylated CSF-1 is administered i.p., s.c. or i.v. to mice at doses up to 4 mg/kg (per mouse) starting up to 24 hours before viral challenge, there is a significant increase in survival as compared to saline- treated control.
Thus, PEGylated CSF-1 may be used alone or in combination witii another lymphokine in the treatment of viral infections in general, and in particular, may be beneficial in immunosuppressive viral infection such as acquired immune deficiency syndrome (AIDS). Example X
In Vivo Stimulation of White Blood Cell Count
Outbred CD- 1 mice are administered purified recombinant human CSF-1 PEGylated as described in Example HI, in amounts up to approximately 2 mg kg per dose on various dosing schedules which are dependent upon die PEGylated CSF-1 preparation used. Total white blood cell count, neutrophil count monocyte count and lymphocyte counts are determined. Increases in any of tiiese parameters may be useful in clinical or veterinary medicine as a stimulus of granulocyte or monocyte production and an enhancer of white blood count
Example XI
PEGylated CSF-1 in Wound Healing
PEGylated CSF-1 is assayed for wound healing using animal models and protocols such as d e Goretex miniature wound healing model of Goodson and Hunt, 1982, J. Surg. Res..22:394, in which implanted Goretex tubes fill up with invading macrophages, fibroblasts and other connective tissue cells, and collagen and fibrin deposition. Healing is assessed by examining tube contents microscopically. A second model is die incisional wound healing model of Eisenger, ei al., 1988, Proc. Nad. Acad. Sci.. USA. £5: 1937, in which wounds are observed visually and punch biopsies are taken to monitor healing, density of cells, and number of epidermal cell layers arising from hair follicles. Also at the end of the experiment wound breaking strength is determined. A third model is a serosal model such as die heat- injured testicular serosa of Fotev, ei al., 1987, J. Pathol..151:209, in which healing is assessed in fixed sections by degree of mesotiielial resurfacing of the injured site. The teachings of each of these models are incorporated herein by reference.
Generally, PEGylated CSF-1 is applied to the site of the wound by soaking a nonadhesive surgical dressing in amounts up to 1,000,000 U/ml of PEGylated CSF-1 in saline under sterile conditions as described in the incisional wound healing model reference using epidermal cell derived factor (EDF) for topical wounds. Altematively, similar amounts of PEGylated CSF-1 are introduced into Goretex tubes at the time of implantation as described in Goodson and Hunt, ibid, or PEGylated CSF- 1 may also be incorporated into a slow-release matrix and applied at die site of the wound (in . Goretex tubes, in or under dressings, or by injection in the serosal cavity) or PEGylated CSF-1 is administered systemically (i.v., i.p., or s.c.) at doses of up to 1,000 μg/kg/day.
The healing rate of each model is measured and tissue repair evaluated in the presence and in the absence of PEGylated CSF-1.
PEGylated CSF-1 may also be used in combination with other growth factors to promote wound healing such as EDF, epidermal growth factor (EGF), fibroblast growth factor (basic and acidic FGF), platelet derived growth factor (PDGF), or transforming growth factors (TGF alpha and beta), IL-1 and otiier substances such as somatomedin C and vitamin C.
Example XIT
Preparation of PEGylated LCSF A. Conjugation
PEG"-11,000 was prepared as described in Example IA. CSF-
1 (LCSF/NV3CV221) was prepared according to fermentation purification and refolding procedures disclosed in PCT Publication No. WO88/08003 published October 20, 1988. 5 mg of LCSF was dialyzed into 20 mM HEPES buffer, pH 7.5, containing 50 mM NaCl. PEG"-11,000 was added at 6-fold molar excess over LCSF dimer. The dry activated PEG was added to die stirring 2 mg/ml LCSF solution at 20βC. Aliquots of the reaction mixture were analyzed every 10 min by size exclusion HPLC (SEC) on Zorbax GF250 (Dupont). The reaction was stopped by addition of a 10-fold excess of ε-aminocaproate over PEG. Table VHI shows NFS 60 bioassay data of die
PEG-LCSF reaction mixtures.
Samples containing 2 mg/ml of M-CSF were diluted into PBS containing 12 mg/ml BSA and assayed in the NFS 60 assay. Values represent the mean of duplicate determination performed on serial dilutions of each sample. TABLE Vm NFS Bioassay of PEG-LCSF/NV3CV221 Sample U/ml CSF-1 Activity starting material 3.65 x 107 10 min + PEG 2.51 x 107
20 min + PEG 3.30 x 107
30 min + PEG 1.68 x 107
Size exclusion analysis of the PEGylation reaction showed two major species ofPEG-CSF after the 30 min reaction. Their sizes were consistent with an approximate derivatization of 1 and 2 moles PEG per mole CSF-1 dimer. The SEC profile after 45 min indicated that die reaction had reached completion at about 30 min.
B. Separation of PEG-LCSF from Unmodified LCSF
The sample was adjusted to pH 8.3 witii 1M Tris-HCl, diafiltered to educe salt concentration to 30 mM and applied to a Bio-Gel® TSK-DEAE-5-PW HPLC 75 x 7.5 mm column. The column was equilibrated in 30 M Tris-HCl, pH8.5, and was developed widi a 40 min gradient of 0 to 0.6 M NaCl. All buffers were prepared with depyrogenated water, and die HPLC system was prewashed with 0.1 M NaOH and with 50% etiianol to remove endotoxin. Fractions off the column were analyzed by SDS-PAGE. PEGylated species eluted earlier than unmodified CSF-1 from the column. Some PEG-CSF also appeared in the column passthrough and this material again did not bind upon reapplication to the column. The unbound fraction also contained released NHS anion.
Other Embodiments
The reaction herein may also be carried out employing polyvinyl alcohol or a polyoxyethylated polyol. An active POG-CSF-1 may be prepared as follows.
Polyoxyethylated glycerol (POG) of molecular weight 5000 may be obtained from Polysciences. To 10 g of POG may be added 2.28 g glutaric anhydride (a 10-fold excess over POG). The mixture may be stirred for two hours at 110°C and cooled. This may be dissolved in 20 ml CHCI3 and poured slowly into 500 ml ether with vigorous stirring. The product may be collected and rinsed witii ether to yield about 90% POG-glutarate product. This product may be reacted witii N-hydroxysuccinimide as described in Example IA to yield d e active ester POG-glutaryl N-hydroxysuccinimide (POG*). Then one of the CSF-1 proteins described above may be reacted with the POG*.
Nitrile-substituted polyvinyl alcohol may also be used to prepare an activated polyvinyl alcohol for conjugation to the CSF-1.
Deposits The following cultures described more fully in PCT
Publication No. WO 86/04607 and in European Patent Publication No. 0272779, were deposited in the Cetus Master Culture Collection (CMCC), 1400 Fifty-Third Street Emeryville, CA 94608 USA and in the American Type Culture Collection (ATCC), 12301 Parklawn Drive, Rockville, MD 20852 USA. The CMCC and ATCC accession numbers and ATCC deposit dates for the deposited samples are:
Figure imgf000051_0001
O/E pPLCSF-17 asp59/CV150 in DG116 3044 67,389 April 14, 1987
Figure imgf000051_0002
The deposits above were made pursuant to a contract between the ATCC and the assignee of this patent application, Cetus Corporation. The contract with ATCC provides for permanent availability of the progeny of these plasmids and die cell line to the public on the issuance of the U.S. patent describing and identifying die deposit or the publications or upon the laying open to the public of any U.S. or foreign patent application, whichever comes first and for availability of the progeny of tiiese plasmids and the cell line to one determined by the U.S. Commissioner of Patents and Trademarks to be entided diereto according to 35 USC §122 and d e Commissioner's rules pursuant thereto (including 37 CFR §1.14 with particular reference to 886 OG 638). The assignee of the present application has agreed that if the plasmids and the cell line on deposit should die or be lost or destroyed when cultivated under suitable conditions, they will be promptly replaced on notification witii a viable culture of the same plasmids and cell line. In summary, the present invention is seen to provide various recombinant CSF-1 molecules selectively derivatized witii water-soluble polymers of different sizes using different chemical linkers. Derivatization of the CSF-1 increases the apparent molecular weight of the CSF-1 and increases its in vivo half-life in the plasma of rats. The derivatization may also increase the solubility of the CSF-1 in aqueous medium at physiological pH and may decrease its immunogenicity by decreasing or eliminating aggregation or by shielding its antigenic determinants.
The foregoing written specification is considered to be sufficient to enable one skilled in d e art to practice the invention. The present invention is not to be limited in scope by the cidtures deposited, since the deposited embodiments are intended as a single illustration of one aspect of the invention, and any cultures that are functionally equivalent are within the scope of this invention. The deposit of materials herein does not constitute an admission mat the written description herein contained is inadequate to enable d e practice of any aspect of d e invention, including d e best mode tiiereof, nor are they to be construed as limiting the scope of the claims to the specific illustrations that they represent Indeed, various modifications of die above- described modes for carrying out the invention that are obvious to those skilled in the field of pharmaceutical formulation or related fields are intended to be within the scope of the following claims.

Claims

Claims
1. A biologically active composition comprising a protein that stimulates the formation of primarily macrophage colonies in the in vitro colony stimulating factor-1 (CSF-1) assay, which protein is covalently conjugated to a water-soluble polymer selected from the group consisting of polyethylene or polypropylene glycol homopolymers, polyoxyethylated polyols, and polyvinyl alcohol, wherein said homopolymer is unsubstituted or substituted at one end witii an alkyl group.
2. The composition of claim 1 wherein the protein is a native human CSF-1.
3. The composition of claim 1 wherein the protein is a recombinant human CSF-1.
4. The composition of claim 3 wherein the protein is recombinandy expressed in bacteria and is a homodimer with predicted amino acid sequence selected from the group consisting of LCSF/CV150 through CV464; tyr59LCSF/CVl50 through CV464; SCSF/CV150 through CV166; asp59SCSF/CV150 through CV166; and the gln52 muteins and NV2 and
NV3 muteins tiiereof, wherein LCSF is coded for by die sequence shown as amino acids 1-522 of Figure 2 and SCSF is coded for by the sequence shown as amino acids 1-224 of Figure 1.
5. The composition of claim 4 wherein the protein is a homodimer with predicted amino acid sequence selected from die group consisting of LCSF/CV150; tyr59LCSF/CV150; LCSF/CV190; tyr59LCSF/CVl90; LCSF/CV191; tyr59LCSF/CVl91; LCSF/CV221; tyr59LCSF/CV221; LCSF/CV223; tyr59LCSF/CV223; LCSF/CV236; tyr59LCSF/CV236; LCSF/CV238; tyr59LCSF/CV238; LCSF/CV249; tyr59LCSF/CV249; LCSF/CV258; tyr59LCSF/CV258; LCSF/CV411; tyr59LCSF/CV411; LCSF/CV464; tyr59LCSF/CV464; SCSF/CV150; SCSF/CV153; SCSF/CV158; the corresponding asp59S CSFs; and the gln52 muteins and NV2 and NV3 muteins thereof.
6. The composition of claim 5 wherein the protein is a homodimer coded for by a sequence selected from die group consisting of LCSF/CV150, LCSF/CV190, LCSF/CV221, tyr59LCSF/CVl50, tyr59LCSF/C 190, tyr59LCSF/CV221, SCSF/CV158, SCSF/CV150, asp59SCSF/CV158; asp59SCSF/CV150;and the NV2 and NV3 muteins thereof.
7. The composition of claim 6 wherein the protein is a homodimer coded for by a sequence selected from the group consisting of
SCSF/CV150, asp59SCSF/CV150, SCSF/NV3CV150, asp59SCSF/NV3CV150, SCSF/NV3CV158, asp59SCSF/NV3CVl58 asp59SCSF/NV2CV150 and asp59SCSF/NV2CV158.
8. The composition of claim 1 wherein the protein is a recombinant heterodimer consisting of one subunit containing a cysteine residue witii a free sulfhydryl group reactive with the polymer.
9. The composition of claim 1 wherein the protein is a murine CSF-1.
10. The composition of claim 1 wherein the protein is a human CSF-1 expressed in and secreted from a eukaryotic host
11. The composition of claim 10 wherein the protein is a dimer coded for by a sequence selected from d e group consisting of
LCSF/CV150; tyr59LCSF/CV150; LCSF/CV190; tyr59LCSF/C l90
LCSF/CV191; tyr59LCSF/CV191; LCSF/CV221; tyr59LCSF/CV221 LCSF/CV223; tyr59LCSF/CV223; LCSF/CV236; tyr59LCSF/CV236
LCSF/CV238; tyr59LCSF/CV238; LCSF/CV249; tyr59LCSF/CV249 LCSF/CV258; tyr59LCSF/CV258; LCSF/CV411; tyr59LCSF/CV411; LCSF/CV464; tyr59LCSF/CV464; SCSF/CV150; SCSF/CV153; SCSF/CV158; the corresponding asps9SCSFs; and the glns2 muteins thereof.
12. The composition of claim 1 wherein said polymer has an average molecular weight of about 1000 to 100,000 daltons.
13. The composition of claim 1 wherein said polymer has an average molecular weight of 4000 to 40,000 daltons.
14. The composition of claim 1 wherein said polymer is conjugated to the protein via reaction with die active ester of a carboxylic acid of said polymer.
15. The composition of claim 14 wherein said polymer is an unsubstituted polyethylene glycol homopolymer, a monomethyl polyethylene glycol homopolymer or a polyoxyethylated glycerol.
16. The composition of claim 1 wherein the protein is conjugated via one to three free amino groups thereof, and d e polymer contains an N-hydroxysuccinimide ester, ] nitrophenyl ester or 4-hydroxy-3- nitrobenzene sulfonic acid ester group tiiat reacts with the free amino groups of the protein.
17. The composition of claim 16 wherein the amino group(s) conjugated is (are) in lysine residue(s) or the N-terminal amino acid or a combination thereof.
18. The composition of claim 1 wherein the protein is conjugated via one or more cysteine residue(s) and the polymer contains a maleimido or haloacetyl group that reacts witii the free sulfhydryl group of the cysteine residue.
19. The composition of claim 1 wherein the protein is glycosylated and is conjugated to d e polymer via at least one of the carbohydrate moieties on the protein, and die polymer contains an amino, hydrazine, or hydrazide group that reacts with free aldehydes formed by oxidation of the carbohydrate moieties.
20. The composition of claim 19 wherein the protein is a dimeric product of a recombinant CSF-1 clone recombinantiy expressed in a eukaryotic host
21. The composition of claim 20 wherein the eukaryotic host is mammalian, insect, yeast or fungal cells.
22. A pharmaceutical preparation comprising the composition of claim 1 dissolved in a pharmaceutically acceptable aqueous carrier medium.
23. The preparation of claim 22 wherein the protein is refolded, and purified in vitro and is die product of a recombinant CSF-1 clone recombinantiy expressed in bacteria.
24. The preparation of claim 23 wherein the protein is a biologically active, refolded CSF-1 dimer having an endotoxin content of less tiian 1.0 ng/mg of CSF-1 and substantially free of pyrogens.
25. The composition of claim 1 wherein said protein is conjugated to said polymer through a urethane bond.
26. The composition of claim 1 wherein said polymer is conjugated to the protein via reaction with the active ester of a carbonic acid of said polymer.
27. The composition of claim 25 wherein said polymer is an unsubstituted polyethylene glycol homopolymer, a monomethyl polyethylene glycol homopolymer or a polyoxyediylated glycerol.
28. The composition of claim 26 wherein there are between 1 and 3 moles polymer, inclusive, per mole CSF-1 dimer.
29. The composition of claim 1 wherein said covalendy conjugated protein is substantially pure witii respect to the number of moles polymer per mole CSF-1.
30. The composition of claim 29 wherein said substantially 5 pure conjugated protein is obtained by ammonium sulfate fractionation.
31. A process for preparing a conjugated protein, which protein stimulates the formation of primarily macrophage colonies in the in vitro colony stimulating factor- 1 (CSF-1) assay, comprising:
(a) providing a water-soluble polymer having at least one o terminal reactive group, where said polymer is selected from the group consisting of polyethylene or polypropylene glycol homopolymers, polyoxyethylated polyols, and polyvinyl alcohol, wherein said homopolymer is unsubstituted or substituted at one end witii an alkyl group;
(b) reacting the protein with the reactive group of said polymer 5 so as to render die polypeptide biologically active and selectively conjugated; and
(c) purifying the conjugated protein.
32. The process of claim 31 wherein said polymer has an average molecular weight of about 1000 to 100,000 daltons.
33. The process of claim 31 wherein die protein is conjugated via one to tiiree free amino groups thereof, and die polymer contains an N- hydroxysuccinimide ester, j>-nitrophenyl ester, or 4-hydroxy-3-nitrobenzene sulfonic acid ester group that reacts with the free amino groups of the protein.
34. The process of claim 33 wherein the amino group(s) is (are) in lysine residue(s) or die N-terminal amino acid or a combination diereof and step (b) is carried out at a pH of about 7-9.
35. The process of claim 31 wherein said polymer is an unsubstituted polyetiiylene glycol homopolymer, a monomethyl polyethylene glycol homopolymer, or a polyoxyethylated glycerol.
36. The process of claim 31 wherein the protein is a recombinant human CSF-1.
37. The process of claim 31 wherein the protein is recombinantiy expressed in bacteria and is a homodimer witii predicted amino acid sequence selected from the group consisting of LCSF/C 150 through CV464; tyr59LCSF/CVl50 through CV464; SCSF/CV150 through CV166; asp59SCSF/CV150 through CV166; and the gln52 muteins and NV2 and
NV3 muteins thereof, wherein LCSF is coded for by the sequence shown as amino acids 1-522 of Figure 2 and SCSF is coded for by the sequence shown as amino acids 1-224 of Figure 1.
38. The process of claim 31 wherein the protein is a human CSF-1 expressed in and secreted from a eukaryotic hos
39. The process of claim 31 further comprising, after step (c), the step of formulating the protein in a pharmaceutically acceptable aqueous carrier medium.
40. The process of claim 31 wherein the protein is glycosylated and is conjugated to die polymer via at least one of the carbohydrate moieties on the protein, and die polymer contains an amino, hydrazine, or hydrazide group that reacts with free aldehydes formed by oxidation of die carbohydrate moieties.
41. The process of claim 40 wherein the protein is a dimeric product of a recombinant CSF-1 clone recombinantiy expressed in a eukaryotic host
42. The process of claim 41 wherein the eukaryotic host is mammalian, yeast, insect, or fungal cells.
43. The process of claim 31 wherein step (c) comprises ammonium sulfate fractionation.
44. A method for prophylactic or tiierapeutic treatment of infectious diseases in mammals comprising administering to said mammal an immunodierapeutically effective amount of the preparation of claim 22.
45. A method for treating tumor burden in mammals comprising administering to said mammal an immunotherapeutically effective amount of the preparation of claim 22.
PCT/US1989/000270 1988-01-20 1989-01-23 Conjugation of polymer to colony stimulating factor-1 WO1989006546A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
DE68913520T DE68913520T2 (en) 1988-01-20 1989-01-23 CONJUGATION OF THE POLYMERS IN COLONIAL STIMULATING FACTOR.
AT89902670T ATE102050T1 (en) 1988-01-20 1989-01-23 CONJUGATION OF POLYMERS TO COLONY STIMULATING FACTOR.

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US146,275 1988-01-20
US07/146,275 US4847325A (en) 1988-01-20 1988-01-20 Conjugation of polymer to colony stimulating factor-1

Publications (1)

Publication Number Publication Date
WO1989006546A1 true WO1989006546A1 (en) 1989-07-27

Family

ID=22516615

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1989/000270 WO1989006546A1 (en) 1988-01-20 1989-01-23 Conjugation of polymer to colony stimulating factor-1

Country Status (9)

Country Link
US (1) US4847325A (en)
EP (1) EP0402378B1 (en)
JP (1) JP3280016B2 (en)
AU (1) AU3180789A (en)
CA (1) CA1340297C (en)
DE (1) DE68913520T2 (en)
IE (1) IE64887B1 (en)
IL (1) IL89001A (en)
WO (1) WO1989006546A1 (en)

Cited By (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1990004606A1 (en) * 1988-10-20 1990-05-03 Royal Free Hospital School Of Medicine A process for fractionating polyethylene glycol (peg)-protein adducts and an adduct of peg and granulocyte-macrophage colony stimulating factor
WO1990007938A1 (en) * 1989-01-23 1990-07-26 Cetus Corporation Preparation of a polymer/interleukin-2 conjugate
WO1990012874A2 (en) * 1989-04-21 1990-11-01 Genetics Institute, Inc. Cysteine added variants of polypeptides and chemical modifications thereof
EP0452505A1 (en) * 1989-11-07 1991-10-23 Otsuka Pharmaceutical Co., Ltd. M-csf derivative, expression vector of said derivative, product of transformation by said vector, and production of them
EP0458064A2 (en) * 1990-05-04 1991-11-27 American Cyanamid Company Stabilization of somatotropins and other proteins by modification of cysteine residues
EP0470128A1 (en) 1989-04-19 1992-02-12 Novo Nordisk A/S Active carbonates of polyalkylene oxides for modification of polypeptides
EP0473268A2 (en) * 1990-07-23 1992-03-04 Zeneca Limited Continuous release pharmaceutical compositions comprising a polypeptide covalently conjugated to a water soluble polymer
EP0473084A2 (en) * 1990-08-28 1992-03-04 Sumitomo Pharmaceuticals Company, Limited Polyethylene glycol derivatives, their modified peptides, methods for producing them and use of the modified peptides
EP0510356A1 (en) * 1991-03-25 1992-10-28 F. Hoffmann-La Roche Ag Polyethylene glycol protein conjugates
EP0539167A2 (en) * 1991-10-21 1993-04-28 Ortho Pharmaceutical Corporation Peg imidates and protein derivatives thereof
WO1994005332A2 (en) * 1992-09-01 1994-03-17 Berlex Laboratories, Inc. Glycolation of glycosylated macromolecules
EP0605963A2 (en) * 1992-12-09 1994-07-13 Ortho Pharmaceutical Corporation Peg hydrazone and peg oxime linkage forming reagents and protein derivatives thereof
US5349052A (en) * 1988-10-20 1994-09-20 Royal Free Hospital School Of Medicine Process for fractionating polyethylene glycol (PEG)-protein adducts and an adduct for PEG and granulocyte-macrophage colony stimulating factor
US5382657A (en) * 1992-08-26 1995-01-17 Hoffmann-La Roche Inc. Peg-interferon conjugates
EP0675201A1 (en) * 1994-03-31 1995-10-04 Amgen Inc. Compositions and methods for stimulating megakaryocyte growth and differentiation
US5650297A (en) * 1986-09-17 1997-07-22 Otsuka Pharmaceutical Co., Ltd. DNA encoding human colony-stimulating factors
EP0790306A2 (en) * 1989-07-18 1997-08-20 Amgen Boulder Inc. Tumor necrosis factor (tnf) inhibitor and use thereof
US5795569A (en) * 1994-03-31 1998-08-18 Amgen Inc. Mono-pegylated proteins that stimulate megakaryocyte growth and differentiation
WO2000013699A1 (en) * 1998-09-04 2000-03-16 Ludwig Institute For Cancer Research An antigenic peptide encoded by an alternative open reading frame of human macrophage colony-stimulating factor
US6083521A (en) * 1993-08-27 2000-07-04 Novartis Ag Polymeric matrices and their uses in pharmaceutical compositions
US6552170B1 (en) 1990-04-06 2003-04-22 Amgen Inc. PEGylation reagents and compounds formed therewith
US6989147B2 (en) 1996-07-09 2006-01-24 Amgen Inc. Truncated soluble tumor necrosis factor type-I and type-II receptors
US7795210B2 (en) 2001-10-10 2010-09-14 Novo Nordisk A/S Protein remodeling methods and proteins/peptides produced by the methods
US7803777B2 (en) 2003-03-14 2010-09-28 Biogenerix Ag Branched water-soluble polymers and their conjugates
US7842661B2 (en) 2003-11-24 2010-11-30 Novo Nordisk A/S Glycopegylated erythropoietin formulations
US7893019B2 (en) 2006-08-11 2011-02-22 Bio-Ker S.R.L. G-CSF site-specific mono-conjugates
US7956032B2 (en) 2003-12-03 2011-06-07 Novo Nordisk A/S Glycopegylated granulocyte colony stimulating factor
US8063015B2 (en) 2003-04-09 2011-11-22 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
US8207112B2 (en) 2007-08-29 2012-06-26 Biogenerix Ag Liquid formulation of G-CSF conjugate
US8268967B2 (en) 2004-09-10 2012-09-18 Novo Nordisk A/S Glycopegylated interferon α
US8361961B2 (en) 2004-01-08 2013-01-29 Biogenerix Ag O-linked glycosylation of peptides
US8633157B2 (en) 2003-11-24 2014-01-21 Novo Nordisk A/S Glycopegylated erythropoietin
US8716239B2 (en) 2001-10-10 2014-05-06 Novo Nordisk A/S Granulocyte colony stimulating factor: remodeling and glycoconjugation G-CSF
US8716240B2 (en) 2001-10-10 2014-05-06 Novo Nordisk A/S Erythropoietin: remodeling and glycoconjugation of erythropoietin
US8791066B2 (en) 2004-07-13 2014-07-29 Novo Nordisk A/S Branched PEG remodeling and glycosylation of glucagon-like peptide-1 [GLP-1]
US8841439B2 (en) 2005-11-03 2014-09-23 Novo Nordisk A/S Nucleotide sugar purification using membranes
US8911967B2 (en) 2005-08-19 2014-12-16 Novo Nordisk A/S One pot desialylation and glycopegylation of therapeutic peptides
US8916360B2 (en) 2003-11-24 2014-12-23 Novo Nordisk A/S Glycopegylated erythropoietin
US8969532B2 (en) 2006-10-03 2015-03-03 Novo Nordisk A/S Methods for the purification of polypeptide conjugates comprising polyalkylene oxide using hydrophobic interaction chromatography
US9005625B2 (en) 2003-07-25 2015-04-14 Novo Nordisk A/S Antibody toxin conjugates
US9029331B2 (en) 2005-01-10 2015-05-12 Novo Nordisk A/S Glycopegylated granulocyte colony stimulating factor
US9050304B2 (en) 2007-04-03 2015-06-09 Ratiopharm Gmbh Methods of treatment using glycopegylated G-CSF
US9125880B2 (en) 2002-12-26 2015-09-08 Mountain View Pharmaceuticals, Inc. Polymer conjugates of interferon-beta with enhanced biological potency
US9150848B2 (en) 2008-02-27 2015-10-06 Novo Nordisk A/S Conjugated factor VIII molecules
US9187546B2 (en) 2005-04-08 2015-11-17 Novo Nordisk A/S Compositions and methods for the preparation of protease resistant human growth hormone glycosylation mutants
US9187532B2 (en) 2006-07-21 2015-11-17 Novo Nordisk A/S Glycosylation of peptides via O-linked glycosylation sequences
US9200049B2 (en) 2004-10-29 2015-12-01 Novo Nordisk A/S Remodeling and glycopegylation of fibroblast growth factor (FGF)
US9493499B2 (en) 2007-06-12 2016-11-15 Novo Nordisk A/S Process for the production of purified cytidinemonophosphate-sialic acid-polyalkylene oxide (CMP-SA-PEG) as modified nucleotide sugars via anion exchange chromatography

Families Citing this family (190)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5837229A (en) * 1985-02-05 1998-11-17 Chiron Corporation Uses of recombinant colony stimulating factor-1
US5422105A (en) * 1985-02-05 1995-06-06 Cetus Oncology Corporation Use of recombinant colony stimulating factor 1
US5556620A (en) * 1985-02-05 1996-09-17 Cetus Oncology Corporation Use of recombinant colony stimulating factor-1 to enhance wound healing
US5573930A (en) * 1985-02-05 1996-11-12 Cetus Oncology Corporation DNA encoding various forms of colony stimulating factor-1
US5104650A (en) * 1985-02-05 1992-04-14 Cetus Corporation Uses of recombinant colony stimulating factor-1
US6117422A (en) * 1985-02-05 2000-09-12 Chiron Corporation N∇2-CSF-1(long form) and carboxy truncated fragments thereof
US6156300A (en) * 1985-02-05 2000-12-05 Chiron Corporation Point mutants of N∇2 CSF-1 and carboxy truncated fragments thereof
US5084559A (en) * 1987-03-27 1992-01-28 Repligen Corporation Protein a domain mutants
US5153265A (en) * 1988-01-20 1992-10-06 Cetus Corporation Conjugation of polymer to colony stimulating factor-1
JPH01193227A (en) * 1988-01-29 1989-08-03 Res Dev Corp Of Japan Adjuvant for cancer immunotherapy
US5322678A (en) * 1988-02-17 1994-06-21 Neorx Corporation Alteration of pharmacokinetics of proteins by charge modification
US5162430A (en) * 1988-11-21 1992-11-10 Collagen Corporation Collagen-polymer conjugates
US5565519A (en) * 1988-11-21 1996-10-15 Collagen Corporation Clear, chemically modified collagen-synthetic polymer conjugates for ophthalmic applications
US5306500A (en) * 1988-11-21 1994-04-26 Collagen Corporation Method of augmenting tissue with collagen-polymer conjugates
US5264214A (en) * 1988-11-21 1993-11-23 Collagen Corporation Composition for bone repair
US5510418A (en) * 1988-11-21 1996-04-23 Collagen Corporation Glycosaminoglycan-synthetic polymer conjugates
US20020177688A1 (en) * 1988-12-22 2002-11-28 Kirin-Amgen, Inc., Chemically-modified G-CSF
US6166183A (en) * 1992-11-30 2000-12-26 Kirin-Amgen, Inc. Chemically-modified G-CSF
DE68925966T2 (en) * 1988-12-22 1996-08-29 Kirin Amgen Inc CHEMICALLY MODIFIED GRANULOCYTE COLONY EXCITING FACTOR
US6225449B1 (en) * 1991-10-04 2001-05-01 Washington University Hormone analogs with multiple CTP extensions
US5288487A (en) * 1989-02-28 1994-02-22 Morinaga Milk Industry Co., Ltd. Human monocyte-macrophage-CSF preparations
US7264944B1 (en) * 1989-04-21 2007-09-04 Amgen Inc. TNF receptors, TNF binding proteins and DNAs coding for them
US7144731B2 (en) * 1989-10-16 2006-12-05 Amgen Inc. SCF antibody compositions and methods of using the same
DE69033584T2 (en) 1989-10-16 2001-03-08 Amgen Inc Stem cell factor
HUT62011A (en) * 1989-10-16 1993-03-29 Amgen Inc Process for producing factors influencing the functioning of stem cells
US20040181044A1 (en) * 1989-10-16 2004-09-16 Zsebo Krisztina M. Method of stimulating growth of epithelial cells by administering stem cell factor
US6852313B1 (en) 1989-10-16 2005-02-08 Amgen Inc. Method of stimulating growth of melanocyte cells by administering stem cell factor
US6204363B1 (en) 1989-10-16 2001-03-20 Amgen Inc. Stem cell factor
US6207454B1 (en) 1989-10-16 2001-03-27 Amgen Inc. Method for enhancing the effciency of gene transfer with stem cell factor (SCF) polypeptide
US6248319B1 (en) 1989-10-16 2001-06-19 Krisztina M. Zsebo Method for increasing hematopoietic progenitor cells by stem cell factor polypeptides
JPH04218000A (en) * 1990-02-13 1992-08-07 Kirin Amgen Inc Modified polypeptide
US5951972A (en) * 1990-05-04 1999-09-14 American Cyanamid Company Stabilization of somatotropins and other proteins by modification of cysteine residues
US5219991A (en) * 1990-09-21 1993-06-15 The United States Of America As Represented By The Department Of Health And Human Services Macrophage stimulating protein
ES2199935T3 (en) * 1991-03-15 2004-03-01 Amgen Inc. PEGILATION OF POLYPEPTIDES.
JPH06506217A (en) * 1991-03-18 1994-07-14 エンゾン,インコーポレーテッド Hydrazine-containing conjugates of polypeptides or glycopolypeptides and polymers
WO1993011431A1 (en) * 1991-11-25 1993-06-10 Baxter Diagnostics Inc. Immunoassay using synthetic htlv-ii peptides
IT1260468B (en) * 1992-01-29 1996-04-09 METHOD FOR MAINTAINING THE ACTIVITY OF PROTEOLYTIC ENZYMES MODIFIED WITH POLYETHYLENGLYCOL
FR2687681B1 (en) * 1992-02-20 1995-10-13 Transgene Sa POLYETHYLENEGLYCOL-HIRUDINE CONJUGATES, THEIR PREPARATION PROCESS AND THEIR USE FOR THE TREATMENT OF THROMBOSES.
EP0648239A4 (en) * 1992-07-02 1995-09-27 Collagen Corp Biocompatible polymer conjugates.
WO1994020453A1 (en) * 1993-03-09 1994-09-15 Enzon, Inc. Taxol polyalkylene oxide conjugates of taxol and taxol intermediates
AU7097094A (en) * 1993-06-01 1994-12-20 Enzon, Inc. Carbohydrate-modified polymer conjugates with erythropoietic activity
AU8052194A (en) * 1993-10-20 1995-05-08 Enzon, Inc. 2'- and/or 7- substituted taxoids
US8192955B1 (en) 1994-01-03 2012-06-05 Genentech, Inc. Nucleic acids encoding MPL ligand (thrombopoietin), variants, and fragments thereof
US8241900B1 (en) 1994-01-03 2012-08-14 Genentech, Inc. mpl ligand
US8147844B1 (en) 1994-01-03 2012-04-03 Genentech, Inc. Mpl ligand (thrombopoietin), nucleic acids encoding such, and methods of treatment using mpl ligand
DE4404168A1 (en) * 1994-02-10 1995-08-17 Hoechst Ag Hirudin derivatives and process for their preparation
US8278099B1 (en) 1994-02-15 2012-10-02 Genentech, Inc. Monoclonal antibody to human thrombopoietin
US20030053982A1 (en) * 1994-09-26 2003-03-20 Kinstler Olaf B. N-terminally chemically modified protein compositions and methods
US5824784A (en) * 1994-10-12 1998-10-20 Amgen Inc. N-terminally chemically modified protein compositions and methods
US6833408B2 (en) * 1995-12-18 2004-12-21 Cohesion Technologies, Inc. Methods for tissue repair using adhesive materials
US7883693B2 (en) 1995-12-18 2011-02-08 Angiodevice International Gmbh Compositions and systems for forming crosslinked biomaterials and methods of preparation of use
US6458889B1 (en) 1995-12-18 2002-10-01 Cohesion Technologies, Inc. Compositions and systems for forming crosslinked biomaterials and associated methods of preparation and use
PT876165E (en) * 1995-12-18 2006-10-31 Angiotech Biomaterials Corp COMPOSITIONS OF RETICULATED POLYMERS AND PROCESSES FOR THEIR USE
US6072040A (en) 1996-10-15 2000-06-06 Medical Analysis Systems, Inc. Stabilized conjugates of uncomplexed subunits of multimeric proteins
PT942740E (en) 1996-12-06 2004-01-30 Amgen Inc COMBINATION THERAPY USING A TNF LIGACTION PROTEIN FOR TREATMENT OF TNF-MEDIATED DISEASES
US8003386B1 (en) * 1997-01-14 2011-08-23 Human Genome Sciences, Inc. Tumor necrosis factor receptors 6α and 6β
US20040013664A1 (en) * 1997-01-14 2004-01-22 Gentz Reiner L. Tumor necrosis factor receptors 6 alpha & 6 beta
US7285267B2 (en) 1997-01-14 2007-10-23 Human Genome Sciences, Inc. Tumor necrosis factor receptors 6α & 6β
US5965704A (en) * 1997-08-05 1999-10-12 Zymogenetics, Inc. Class two cytokine receptor-11
US6391311B1 (en) * 1998-03-17 2002-05-21 Genentech, Inc. Polypeptides having homology to vascular endothelial cell growth factor and bone morphogenetic protein 1
US6458355B1 (en) * 1998-01-22 2002-10-01 Genentech, Inc. Methods of treating inflammatory disease with anti-IL-8 antibody fragment-polymer conjugates
US6468532B1 (en) * 1998-01-22 2002-10-22 Genentech, Inc. Methods of treating inflammatory diseases with anti-IL-8 antibody fragment-polymer conjugates
US7371836B2 (en) * 1998-03-27 2008-05-13 Genentech, Inc. PRO526 nucleic acids
PT1588716E (en) * 1998-08-06 2011-05-25 Mountain View Pharmaceuticals Peg-urate oxidase conjugates and use thereof
US20060188971A1 (en) * 1998-08-06 2006-08-24 Duke University Urate oxidase
WO2000008196A2 (en) 1998-08-06 2000-02-17 Duke University Urate oxidase
US6783965B1 (en) 2000-02-10 2004-08-31 Mountain View Pharmaceuticals, Inc. Aggregate-free urate oxidase for preparation of non-immunogenic polymer conjugates
PT1121382E (en) * 1998-10-16 2006-10-31 Biogen Idec Inc BETA INTERFERENCE FUSE PROTEINS AND THEIR RESPECTIVE USES
PT1121156E (en) 1998-10-16 2006-05-31 Biogen Idec Inc CONJUGATES OF INTERFERRO-BETA-1A POLYMERS AND THEIR USES
US6660843B1 (en) * 1998-10-23 2003-12-09 Amgen Inc. Modified peptides as therapeutic agents
WO2000028035A1 (en) * 1998-11-10 2000-05-18 Human Genome Sciences Inc. CHEMOKINE β-7
KR100689212B1 (en) * 1999-01-29 2007-03-09 암겐 인코포레이티드 Gcsf conjugates
CA2369869C (en) * 1999-04-02 2008-06-10 Ajinomoto Co., Inc. Process for producing subunit peptide originating in polymer protein
US20050079577A1 (en) * 1999-05-14 2005-04-14 Ashkenazi Avi J. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US6514500B1 (en) * 1999-10-15 2003-02-04 Conjuchem, Inc. Long lasting synthetic glucagon like peptide {GLP-!}
DE60139110D1 (en) 2000-01-10 2009-08-13 Maxygen Holdings Ltd G-CSF CONJUGATES
DK1257295T3 (en) 2000-02-11 2009-08-10 Bayer Healthcare Llc Factor VII or VIIA-like molecules
AU2002216749A1 (en) * 2000-06-30 2002-01-14 Smithkline Beecham Corporation Chemokine conjugates
AU2001290524A1 (en) * 2000-08-08 2002-02-18 Zymogenetics Inc. Soluble zcytor 11 cytokine receptors
EP1330654B1 (en) * 2000-10-23 2006-03-08 Genetics Institute, LLC Acid-labile isotope-coded extractant (alice) in mass spectometric analysis
EP1353622A4 (en) * 2000-11-03 2005-06-22 Smithkline Beecham Corp Method of improving systemic exposure of subcutaneously administered therapeutic proteins
DE60232434D1 (en) 2001-02-27 2009-07-09 Maxygen Aps NEW INTERFERON-BETA-LIKE MOLECULES
DE10112825A1 (en) * 2001-03-16 2002-10-02 Fresenius Kabi De Gmbh HESylation of active ingredients in aqueous solution
CA2446942C (en) * 2001-05-16 2010-07-20 Yeda Research And Development Co., Ltd. Use of il-18 inhibitors for the treatment or prevention of sepsis
US20020198139A1 (en) * 2001-05-17 2002-12-26 Deutschman Clifford S. Method of preventing acute pulmonary cell injury
ES2556338T3 (en) * 2001-10-10 2016-01-15 Novo Nordisk A/S Remodeling and glycoconjugation of peptides
US7157277B2 (en) * 2001-11-28 2007-01-02 Neose Technologies, Inc. Factor VIII remodeling and glycoconjugation of Factor VIII
US8008252B2 (en) * 2001-10-10 2011-08-30 Novo Nordisk A/S Factor VII: remodeling and glycoconjugation of Factor VII
DE10209821A1 (en) * 2002-03-06 2003-09-25 Biotechnologie Ges Mittelhesse Coupling of proteins to a modified polysaccharide
DE10209822A1 (en) * 2002-03-06 2003-09-25 Biotechnologie Ges Mittelhesse Coupling of low molecular weight substances to a modified polysaccharide
AU2003241279A1 (en) * 2002-03-26 2003-10-13 Biosynexus Incorporated Antimicrobial polymer conjugates
JP2006507850A (en) * 2002-03-26 2006-03-09 バイオシネクサス インコーポレイテッド Enzymatic destruction of bacterial biofilms
PL211175B1 (en) 2002-06-21 2012-04-30 Novo Nordisk Healthcare Ag Pegylated factor vii glycoforms
DE60336555D1 (en) 2002-06-21 2011-05-12 Novo Nordisk Healthcare Ag PEGYLATED GLYCO FORMS OF FACTOR VII
CA2497777A1 (en) * 2002-09-05 2004-03-18 The General Hospital Corporation Modified asialo-interferons and uses thereof
WO2004024776A1 (en) * 2002-09-11 2004-03-25 Fresenius Kabi Deutschland Gmbh Method of producing hydroxyalkyl starch derivatives
DE02020425T1 (en) * 2002-09-11 2004-07-15 Fresenius Kabi Deutschland Gmbh Hasylated polypeptides, especially hasylated erythropoietin
EP1681303B1 (en) * 2002-09-11 2013-09-04 Fresenius Kabi Deutschland GmbH HASylated polypeptides, especially HASylated erythropoietin
DE10242076A1 (en) * 2002-09-11 2004-03-25 Fresenius Kabi Deutschland Gmbh New covalently bonded conjugates of hydroxyalkyl starch with allergens, useful as modified allergens with depot effect for use in specific immunotherapy for combating allergies, e.g. hay fever
WO2004026251A2 (en) * 2002-09-20 2004-04-01 Pharmacia Corporation Process for decreasing aggregate levels of pegylated protein
AU2003273413A1 (en) * 2002-10-08 2004-05-04 Fresenius Kabi Deutschland Gmbh Pharmaceutically active oligosaccharide conjugates
JP2004196770A (en) * 2002-10-24 2004-07-15 Effector Cell Institute Inc Agent for increasing blood level of dendritic cell precursor
CA2511815A1 (en) * 2002-12-26 2004-07-22 Mountain View Pharmaceuticals, Inc. Polymer conjugates of cytokines, chemokines, growth factors, polypeptide hormones and antagonists thereof with preserved receptor-binding activity
WO2004060405A2 (en) * 2002-12-30 2004-07-22 Angiotech International Ag Tissue reactive compounds and compositions and uses thereof
ATE457716T1 (en) 2002-12-30 2010-03-15 Angiotech Int Ag RELEASE OF ACTIVE INGREDIENTS FROM QUICK-GELLING POLYMER COMPOSITION
EP1606317B1 (en) * 2003-03-24 2010-07-28 ZymoGenetics, Inc. Anti-il-22ra antibodies and binding partners and methods of using in inflammation
US20100221237A1 (en) * 2003-03-26 2010-09-02 Biosynexus Incorporated Enzyme disruption of bacterial biofilms
US8791070B2 (en) 2003-04-09 2014-07-29 Novo Nordisk A/S Glycopegylated factor IX
WO2004091517A2 (en) 2003-04-15 2004-10-28 Smithkline Beecham Corporation Conjugates comprising human il-18 and substitution mutants thereof
ES2380093T3 (en) * 2003-05-09 2012-05-08 Biogenerix Ag Compositions and methods for the preparation of human growth hormone glycosylation mutants
WO2005014655A2 (en) * 2003-08-08 2005-02-17 Fresenius Kabi Deutschland Gmbh Conjugates of hydroxyalkyl starch and a protein
US20080274948A1 (en) * 2003-08-08 2008-11-06 Fresenius Kabi Deutschland Gmbh Conjugates of Hydroxyalkyl Starch and G-Csf
EP1653991A2 (en) * 2003-08-08 2006-05-10 Fresenius Kabi Deutschland GmbH Conjugates of a polymer and a protein linked by an oxime linking group
WO2005035565A1 (en) 2003-10-10 2005-04-21 Novo Nordisk A/S Il-21 derivatives
ES2428358T3 (en) 2003-10-17 2013-11-07 Novo Nordisk A/S Combination therapy
US20050208095A1 (en) * 2003-11-20 2005-09-22 Angiotech International Ag Polymer compositions and methods for their use
US20060040856A1 (en) * 2003-12-03 2006-02-23 Neose Technologies, Inc. Glycopegylated factor IX
AU2005319099B2 (en) 2004-02-02 2010-09-16 Ambrx, Inc. Modified human growth hormone
WO2005074524A2 (en) 2004-02-02 2005-08-18 Ambrx, Inc. Modified human interferon polypeptides and their uses
TWI417303B (en) * 2004-03-11 2013-12-01 Fresenius Kabi De Gmbh Conjugates of hydroxyalkyl starch and a protein, prepared by reductive amination
AR048918A1 (en) * 2004-03-11 2006-06-14 Fresenius Kabi De Gmbh CONJUGADOS DE ALMIDON DE HIDROXIETILO Y ERITROPOYETINA
NZ550964A (en) 2004-04-28 2011-05-27 Angiodevice Internat Gmbh Compositions and systems for forming crosslinked biomaterials and associated methods of preparation and use
KR101699142B1 (en) 2004-06-18 2017-01-23 암브룩스, 인코포레이티드 Novel antigen-binding polypeptides and their uses
US20090292110A1 (en) * 2004-07-23 2009-11-26 Defrees Shawn Enzymatic modification of glycopeptides
WO2006034128A2 (en) 2004-09-17 2006-03-30 Angiotech Biomaterials Corporation Multifunctional compounds for forming crosslinked biomaterials and methods of preparation and use
KR101238684B1 (en) * 2004-10-22 2013-03-04 지모제넥틱스, 인코포레이티드 Anti-il-22ra antibodies and binding partners and methods of using in inflammation
US7612153B2 (en) * 2004-10-25 2009-11-03 Intezyne Technologies, Inc. Heterobifunctional poly(ethylene glycol) and uses thereof
CA2592040A1 (en) * 2004-12-22 2006-06-29 Kuros Biosurgery Ag Michael-type addition reaction functionalised peg hydrogels with factor xiiia incorporated biofactors
CN101119743B (en) * 2005-01-31 2012-09-26 株式会社Eci Immunopotentiating agent
AU2006222187A1 (en) * 2005-03-11 2006-09-14 Fresenius Kabi Deutschland Gmbh Production of bioactive glycoproteins from inactive starting material by conjugation with hydroxyalkylstarch
US8148123B2 (en) * 2005-04-11 2012-04-03 Savient Pharmaceuticals, Inc. Methods for lowering elevated uric acid levels using intravenous injections of PEG-uricase
US7811800B2 (en) 2005-04-11 2010-10-12 Savient Pharmaceuticals, Inc. Variant form of urate oxidase and use thereof
JP2008535500A (en) 2005-04-11 2008-09-04 サビエント ファーマセウティカルズ インク. Mutant urate oxidase and use thereof
US20080159976A1 (en) * 2005-04-11 2008-07-03 Jacob Hartman Methods for lowering elevated uric acid levels using intravenous injections of PEG-uricase
US7833979B2 (en) * 2005-04-22 2010-11-16 Amgen Inc. Toxin peptide therapeutic agents
US20090081122A1 (en) * 2005-05-23 2009-03-26 Universite De Geneve Injectable superparamagnetic nanoparticles for treatment by hyperthermia and use for forming an hyperthermic implant
JP5216580B2 (en) * 2005-05-25 2013-06-19 ノヴォ ノルディスク アー/エス Glycopegylated factor IX
ES2553160T3 (en) 2005-06-17 2015-12-04 Novo Nordisk Health Care Ag Selective reduction and derivatization of engineered Factor VII proteins comprising at least one non-native cysteine
US8008453B2 (en) 2005-08-12 2011-08-30 Amgen Inc. Modified Fc molecules
US20090305967A1 (en) * 2005-08-19 2009-12-10 Novo Nordisk A/S Glycopegylated factor vii and factor viia
EP1762250A1 (en) * 2005-09-12 2007-03-14 Fresenius Kabi Deutschland GmbH Conjugates of hydroxyalkyl starch and an active substance, prepared by chemical ligation via thiazolidine
US8168592B2 (en) * 2005-10-21 2012-05-01 Amgen Inc. CGRP peptide antagonists and conjugates
CA2626522A1 (en) 2005-11-16 2007-05-24 Ambrx, Inc. Methods and compositions comprising non-natural amino acids
CN101622270B (en) * 2006-04-12 2014-01-01 萨文特医药公司 Method for purification of proteins with cationic surfactant
US7560588B2 (en) * 2006-04-27 2009-07-14 Intezyne Technologies, Inc. Poly(ethylene glycol) containing chemically disparate endgroups
CN101484576A (en) 2006-05-24 2009-07-15 诺沃-诺迪斯克保健股份有限公司 Factor IX analogues having prolonged in vivo half life
DK2054434T3 (en) * 2006-08-03 2017-07-03 Myostin Therapeutics Pty Ltd Myostatin antagonists
CN106008699A (en) 2006-09-08 2016-10-12 Ambrx公司 Modified human plasma polypeptide or Fc scaffolds and their uses
AU2007343796A1 (en) * 2006-10-25 2008-07-24 Amgen Inc. Toxin peptide therapeutic agents
CN107501407B (en) 2007-03-30 2022-03-18 Ambrx公司 Modified FGF-21 polypeptides and uses thereof
ES2381639T3 (en) 2007-04-13 2012-05-30 Kuros Biosurgery Ag Polymeric fabric sealant
WO2008153745A2 (en) 2007-05-22 2008-12-18 Amgen Inc. Compositions and methods for producing bioactive fusion proteins
US7968811B2 (en) * 2007-06-29 2011-06-28 Harley-Davidson Motor Company Group, Inc. Integrated ignition and key switch
NZ603812A (en) 2007-11-20 2014-06-27 Ambrx Inc Modified insulin polypeptides and their uses
EP2070951A1 (en) * 2007-12-14 2009-06-17 Fresenius Kabi Deutschland GmbH Method for producing a hydroxyalkyl starch derivatives with two linkers
EP2070950A1 (en) * 2007-12-14 2009-06-17 Fresenius Kabi Deutschland GmbH Hydroxyalkyl starch derivatives and process for their preparation
NZ620606A (en) 2008-02-08 2015-08-28 Ambrx Inc Modified leptin polypeptides and their uses
PE20110426A1 (en) 2008-07-23 2011-07-01 Ambrx Inc MODIFIED BOVINE G-CSF POLYPEPTIDES
AU2009284840B2 (en) * 2008-08-25 2012-09-27 Biopolymed Inc. Biopolymer conjugates comprising an interleukin-11 analog
CN107022020A (en) 2008-09-26 2017-08-08 Ambrx公司 The animal erythropoietin polypeptides and its purposes of modification
CN102224238B (en) 2008-09-26 2015-06-10 Ambrx公司 Non-natural amino acid replication-dependent microorganisms and vaccines
SG176897A1 (en) 2009-06-25 2012-01-30 Savient Pharmaceuticals Inc Methods and kits for predicting infusion reaction risk and antibody-mediated loss of response by monitoring serum uric acid during pegylated uricase therapy
NZ623810A (en) * 2009-07-27 2015-10-30 Lipoxen Technologies Ltd Glycopolysialylation of non-blood coagulation proteins
AU2010277438B2 (en) 2009-07-27 2015-08-20 Baxalta GmbH Glycopolysialylation of non-blood coagulation proteins
CA2784793A1 (en) 2009-12-21 2011-07-21 Ambrx, Inc. Modified bovine somatotropin polypeptides and their uses
CA2784800A1 (en) 2009-12-21 2011-07-21 Ambrx, Inc. Modified porcine somatotropin polypeptides and their uses
CA2791841C (en) 2010-03-05 2023-01-03 Rigshospitalet Chimeric inhibitor molecules of complement activation
JP2013528374A (en) 2010-05-10 2013-07-11 パーシード セラピューティクス リミテッド ライアビリティ カンパニー Polypeptide inhibitors of VLA4
LT2601214T (en) * 2010-08-06 2018-02-26 Genzyme Corporation Vegf antagonist compositions and uses thereof
US9567386B2 (en) 2010-08-17 2017-02-14 Ambrx, Inc. Therapeutic uses of modified relaxin polypeptides
RS59193B1 (en) 2010-08-17 2019-10-31 Ambrx Inc Modified relaxin polypeptides and their uses
AR083006A1 (en) 2010-09-23 2013-01-23 Lilly Co Eli FORMULATIONS FOR THE STIMULATING FACTOR OF COLONIES OF GRANULOCITS (G-CSF) BOVINE AND VARIANTS OF THE SAME
AP2013007173A0 (en) 2011-03-16 2013-10-31 Amgen Inc Potent and selective inhibitors of NAV1.3 and NAV1.7
AU2012280474A1 (en) 2011-07-01 2014-01-16 Bayer Intellectual Property Gmbh Relaxin fusion polypeptides and uses thereof
CA2875989A1 (en) 2012-06-08 2013-12-12 Sutro Biopharma, Inc. Antibodies comprising site-specific non-natural amino acid residues, methods of their preparation and methods of their use
DK2863955T3 (en) 2012-06-26 2017-01-23 Sutro Biopharma Inc MODIFIED FC PROTEINS, INCLUDING LOCATION-SPECIFIC NON-NATURAL AMINO ACID RESIDUES, CONJUGATES THEREOF, METHODS OF PRODUCING ITS AND PROCEDURES FOR USE THEREOF
HUE045227T2 (en) 2012-08-31 2019-12-30 Sutro Biopharma Inc Modified amino acids comprising an azido group
US20160067347A1 (en) 2012-12-20 2016-03-10 Amgen Inc. Apj receptor agonists and uses thereof
TW201446792A (en) 2013-03-12 2014-12-16 Amgen Inc Potent and selective inhibitors of Nav1.7
US9764039B2 (en) 2013-07-10 2017-09-19 Sutro Biopharma, Inc. Antibodies comprising multiple site-specific non-natural amino acid residues, methods of their preparation and methods of their use
WO2015054658A1 (en) 2013-10-11 2015-04-16 Sutro Biopharma, Inc. Modified amino acids comprising tetrazine functional groups, methods of preparation, and methods of their use
AU2015274574B2 (en) 2014-06-10 2019-10-10 Amgen Inc. Apelin polypeptides
PT3412302T (en) 2014-10-24 2021-06-09 Bristol Myers Squibb Co Modified fgf-21 polypeptides and uses thereof
CN110637027A (en) 2017-02-08 2019-12-31 百时美施贵宝公司 Modified relaxin polypeptides comprising pharmacokinetic enhancers and uses thereof
SG11202102427XA (en) 2018-09-11 2021-04-29 Ambrx Inc Interleukin-2 polypeptide conjugates and their uses
EP3867265A1 (en) 2018-10-19 2021-08-25 Ambrx, Inc. Interleukin-10 polypeptide conjugates, dimers thereof, and their uses
WO2020168017A1 (en) 2019-02-12 2020-08-20 Ambrx, Inc. Compositions containing, methods and uses of antibody-tlr agonist conjugates
WO2021183832A1 (en) 2020-03-11 2021-09-16 Ambrx, Inc. Interleukin-2 polypeptide conjugates and methods of use thereof
EP4199968A1 (en) 2020-08-20 2023-06-28 Ambrx, Inc. Antibody-tlr agonist conjugates, methods and uses thereof
CA3213805A1 (en) 2021-04-03 2022-10-06 Feng Tian Anti-her2 antibody-drug conjugates and uses thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0098110A2 (en) * 1982-06-24 1984-01-11 NIHON CHEMICAL RESEARCH KABUSHIKI KAISHA also known as JAPAN CHEMICAL RESEARCH CO., LTD Long-acting composition
EP0154316A2 (en) * 1984-03-06 1985-09-11 Takeda Chemical Industries, Ltd. Chemically modified lymphokine and production thereof
WO1986004607A1 (en) * 1985-02-05 1986-08-14 Cetus Corporation Recombinant colony stimulating factor-1
WO1987006954A1 (en) * 1986-05-06 1987-11-19 Genetics Institute, Inc. Production of m-csf
GB2193631A (en) * 1986-07-18 1988-02-17 Chugai Pharmaceutical Co Ltd Stable granulocyte colony stimulating factor composition

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3619371A (en) * 1967-07-03 1971-11-09 Nat Res Dev Production of a polymeric matrix having a biologically active substance bound thereto
SE343210B (en) * 1967-12-20 1972-03-06 Pharmacia Ab
US3876501A (en) * 1973-05-17 1975-04-08 Baxter Laboratories Inc Binding enzymes to activated water-soluble carbohydrates
GB1479268A (en) * 1973-07-05 1977-07-13 Beecham Group Ltd Pharmaceutical compositions
US4179337A (en) * 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
DE2360794C2 (en) * 1973-12-06 1984-12-06 Hoechst Ag, 6230 Frankfurt Process for the production of peptides
CH596313A5 (en) * 1975-05-30 1978-03-15 Battelle Memorial Institute
US4002531A (en) * 1976-01-22 1977-01-11 Pierce Chemical Company Modifying enzymes with polyethylene glycol and product produced thereby
GB1578348A (en) * 1976-08-17 1980-11-05 Pharmacia Ab Products and a method for the therapeutic suppression of reaginic antibodies responsible for common allergic
US4430260A (en) * 1979-02-27 1984-02-07 Lee Weng Y Penicillin-polyvinyl alcohol conjugate and process of preparation
US4296097A (en) * 1979-02-27 1981-10-20 Lee Weng Y Suppression of reaginic antibodies to drugs employing polyvinyl alcohol as carrier therefor
JPS6023084B2 (en) * 1979-07-11 1985-06-05 味の素株式会社 blood substitute
US4415665A (en) * 1980-12-12 1983-11-15 Pharmacia Fine Chemicals Ab Method of covalently binding biologically active organic substances to polymeric substances
JPS6030654B2 (en) * 1980-12-31 1985-07-17 株式会社林原生物化学研究所 Method for producing human colony stimulating factor
JPS59137417A (en) * 1983-01-28 1984-08-07 Morinaga Milk Ind Co Ltd Preparation of colonization stimulation factor and kallikrein originated from human urine
US4504586A (en) * 1983-02-03 1985-03-12 Amgen Hybridoma tumor cell lines and their monoclonal antibodies to human colony stimulating factor subclass number 1
JPS59169489A (en) * 1983-03-15 1984-09-25 Otsuka Pharmaceut Co Ltd Cultured and established human cell
DE3340592A1 (en) * 1983-11-10 1985-05-23 B. Braun Melsungen Ag, 3508 Melsungen CONJUGATES OF MACROMOLECULAR COMPOUNDS TO HAEMOGLOBINE, METHOD FOR THEIR PRODUCTION AND MEDICINAL PRODUCTS CONTAINING THE SAME
US4496689A (en) * 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
US4732863A (en) * 1984-12-31 1988-03-22 University Of New Mexico PEG-modified antibody with reduced affinity for cell surface Fc receptors
US4766106A (en) * 1985-06-26 1988-08-23 Cetus Corporation Solubilization of proteins for pharmaceutical compositions using polymer conjugation
WO1987000056A1 (en) * 1985-06-26 1987-01-15 Cetus Corporation Solubilization of proteins for pharmaceutical compositions using polymer conjugation

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0098110A2 (en) * 1982-06-24 1984-01-11 NIHON CHEMICAL RESEARCH KABUSHIKI KAISHA also known as JAPAN CHEMICAL RESEARCH CO., LTD Long-acting composition
EP0154316A2 (en) * 1984-03-06 1985-09-11 Takeda Chemical Industries, Ltd. Chemically modified lymphokine and production thereof
WO1986004607A1 (en) * 1985-02-05 1986-08-14 Cetus Corporation Recombinant colony stimulating factor-1
WO1987006954A1 (en) * 1986-05-06 1987-11-19 Genetics Institute, Inc. Production of m-csf
GB2193631A (en) * 1986-07-18 1988-02-17 Chugai Pharmaceutical Co Ltd Stable granulocyte colony stimulating factor composition

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
PROC. NATL. ACAD. SCI. U.S.A., Vol. 84, 1987, (USA), NANDINI V. KATRE et al., "Chemical Modification of Recombinant Interleukin 2 by Polyethylene Glycol Increases its Potency in the Murine Meth A Sarcoma Model", pages 1487-1491. *
SCIENCE, Vol. 229, 1985, DONALD METCALF, "The Granulocyte-Macrophage Colony-Stimulating Factors", pages 16-22. *

Cited By (82)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5650297A (en) * 1986-09-17 1997-07-22 Otsuka Pharmaceutical Co., Ltd. DNA encoding human colony-stimulating factors
WO1990004606A1 (en) * 1988-10-20 1990-05-03 Royal Free Hospital School Of Medicine A process for fractionating polyethylene glycol (peg)-protein adducts and an adduct of peg and granulocyte-macrophage colony stimulating factor
US6384195B1 (en) 1988-10-20 2002-05-07 Polymasc Pharmaceuticals Plc. Process for fractionating polyethylene glycol (PEG) —protein adducts and an adduct of PEG and granulocyt-macrophage colony stimulating factor
US5880255A (en) * 1988-10-20 1999-03-09 Polymasc Pharmaceuticals Plc Process for fractionating polyethylene glycol (PEG)-protein adducts and an adduct of PEG and granulocyte-macrophage colony stimulating factor
EP0727438A2 (en) * 1988-10-20 1996-08-21 PolyMASC Pharmaceuticals plc Adducts of PEG and GM-CSF
US5349052A (en) * 1988-10-20 1994-09-20 Royal Free Hospital School Of Medicine Process for fractionating polyethylene glycol (PEG)-protein adducts and an adduct for PEG and granulocyte-macrophage colony stimulating factor
WO1990007938A1 (en) * 1989-01-23 1990-07-26 Cetus Corporation Preparation of a polymer/interleukin-2 conjugate
EP0470128B2 (en) 1989-04-19 2003-08-13 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
EP0470128A1 (en) 1989-04-19 1992-02-12 Novo Nordisk A/S Active carbonates of polyalkylene oxides for modification of polypeptides
EP0893439B1 (en) * 1989-04-19 2005-07-27 Enzon, Inc. A process for forming a modified polypeptide comprising a polypeptide and a polyalkylene oxide
EP0668353A1 (en) * 1989-04-21 1995-08-23 Genetics Institute, Inc. Cysteine added variants of EPO and chemical modifications thereof
EP0668354A1 (en) * 1989-04-21 1995-08-23 Genetics Institute, Inc. Cysteine added variants of G-CSF and chemical modifications thereof
WO1990012874A3 (en) * 1989-04-21 1991-01-10 Genetics Inst Cysteine added variants of polypeptides and chemical modifications thereof
WO1990012874A2 (en) * 1989-04-21 1990-11-01 Genetics Institute, Inc. Cysteine added variants of polypeptides and chemical modifications thereof
EP0790306A2 (en) * 1989-07-18 1997-08-20 Amgen Boulder Inc. Tumor necrosis factor (tnf) inhibitor and use thereof
EP0790306A3 (en) * 1989-07-18 1998-07-01 Amgen Boulder Inc. Tumor necrosis factor (tnf) inhibitor and use thereof
EP0452505A1 (en) * 1989-11-07 1991-10-23 Otsuka Pharmaceutical Co., Ltd. M-csf derivative, expression vector of said derivative, product of transformation by said vector, and production of them
US6552170B1 (en) 1990-04-06 2003-04-22 Amgen Inc. PEGylation reagents and compounds formed therewith
EP0458064A3 (en) * 1990-05-04 1992-06-17 American Cyanamid Company Stabilization of somatotropins and other proteins by modification of cysteine residues
EP0458064A2 (en) * 1990-05-04 1991-11-27 American Cyanamid Company Stabilization of somatotropins and other proteins by modification of cysteine residues
EP0473268A3 (en) * 1990-07-23 1992-09-16 Imperial Chemical Industries Plc Continuous release pharmaceutical compositions comprising a polypeptide covalently conjugated to a water soluble polymer
US5320840A (en) * 1990-07-23 1994-06-14 Imperial Chemical Industries Plc Continuous release pharmaceutical compositions
US5773581A (en) * 1990-07-23 1998-06-30 Zeneca Limited Conjugate of a solution stable G-CSF derivative and a water-soluble polymer
EP0473268A2 (en) * 1990-07-23 1992-03-04 Zeneca Limited Continuous release pharmaceutical compositions comprising a polypeptide covalently conjugated to a water soluble polymer
EP0473084A3 (en) * 1990-08-28 1992-04-29 Sumitomo Pharmaceuticals Company, Limited Polyethylene glycol derivatives, their modified peptides, methods for producing them and use of the modified peptides
US5183660A (en) * 1990-08-28 1993-02-02 Sumitomo Pharmaceuticals Company, Limited Polyethylene glycol derivatives, their modified peptides, methods for producing them and use of the modified peptides
EP0473084A2 (en) * 1990-08-28 1992-03-04 Sumitomo Pharmaceuticals Company, Limited Polyethylene glycol derivatives, their modified peptides, methods for producing them and use of the modified peptides
US5849860A (en) * 1991-03-25 1998-12-15 Hoffmann-La Roche Inc. Polyethylene-protein conjugates
US5539063A (en) * 1991-03-25 1996-07-23 Hoffmann-La Roche Inc. Polymer for making poly(ethylene glycol)-protein conjugates
US5559213A (en) * 1991-03-25 1996-09-24 Hoffmann-La Roche Inc. Polyethylene-protein conjugates
US5595732A (en) * 1991-03-25 1997-01-21 Hoffmann-La Roche Inc. Polyethylene-protein conjugates
US5834594A (en) * 1991-03-25 1998-11-10 Hoffman-La Roche Inc. Polyethylene-protein conjugates
EP0510356A1 (en) * 1991-03-25 1992-10-28 F. Hoffmann-La Roche Ag Polyethylene glycol protein conjugates
US5747646A (en) * 1991-03-25 1998-05-05 Hoffmann-La Roche Inc. Polyethylene-protein conjugates
US5792834A (en) * 1991-03-25 1998-08-11 Hoffmann-La Roche Inc. Polyethylene-protein conjugates
EP0539167A3 (en) * 1991-10-21 1993-08-04 Ortho Pharmaceutical Corporation Peg imidates and protein derivatives thereof
EP0539167A2 (en) * 1991-10-21 1993-04-28 Ortho Pharmaceutical Corporation Peg imidates and protein derivatives thereof
US5382657A (en) * 1992-08-26 1995-01-17 Hoffmann-La Roche Inc. Peg-interferon conjugates
WO1994005332A3 (en) * 1992-09-01 1994-04-14 Berlex Lab Glycolation of glycosylated macromolecules
WO1994005332A2 (en) * 1992-09-01 1994-03-17 Berlex Laboratories, Inc. Glycolation of glycosylated macromolecules
EP0605963A3 (en) * 1992-12-09 1995-11-08 Ortho Pharma Corp Peg hydrazone and peg oxime linkage forming reagents and protein derivatives thereof.
EP0605963A2 (en) * 1992-12-09 1994-07-13 Ortho Pharmaceutical Corporation Peg hydrazone and peg oxime linkage forming reagents and protein derivatives thereof
US6083521A (en) * 1993-08-27 2000-07-04 Novartis Ag Polymeric matrices and their uses in pharmaceutical compositions
US6262127B1 (en) 1993-08-27 2001-07-17 Novartis Ag Polymeric matrices and their uses in pharmaceutical compositions
EP0755263A4 (en) * 1994-03-31 2005-02-09 Amgen Inc Compositions and methods for stimulating megakaryocyte growth and differentiation
EP0675201A1 (en) * 1994-03-31 1995-10-04 Amgen Inc. Compositions and methods for stimulating megakaryocyte growth and differentiation
US5766581A (en) * 1994-03-31 1998-06-16 Amgen Inc. Method for treating mammals with monopegylated proteins that stimulates megakaryocyte growth and differentiation
EP0755263A1 (en) * 1994-03-31 1997-01-29 Amgen Inc. Compositions and methods for stimulating megakaryocyte growth and differentiation
EP0690127A1 (en) * 1994-03-31 1996-01-03 Amgen Inc. Compositions and methods for stimulating megakaryocyte growth and differentiation
US5795569A (en) * 1994-03-31 1998-08-18 Amgen Inc. Mono-pegylated proteins that stimulate megakaryocyte growth and differentiation
US6989147B2 (en) 1996-07-09 2006-01-24 Amgen Inc. Truncated soluble tumor necrosis factor type-I and type-II receptors
US7732587B2 (en) 1996-07-09 2010-06-08 Amgen Inc. Nucleic acids encoding truncated soluble tumor necrosis factor
WO2000013699A1 (en) * 1998-09-04 2000-03-16 Ludwig Institute For Cancer Research An antigenic peptide encoded by an alternative open reading frame of human macrophage colony-stimulating factor
US7795210B2 (en) 2001-10-10 2010-09-14 Novo Nordisk A/S Protein remodeling methods and proteins/peptides produced by the methods
US8716240B2 (en) 2001-10-10 2014-05-06 Novo Nordisk A/S Erythropoietin: remodeling and glycoconjugation of erythropoietin
US8716239B2 (en) 2001-10-10 2014-05-06 Novo Nordisk A/S Granulocyte colony stimulating factor: remodeling and glycoconjugation G-CSF
US9125880B2 (en) 2002-12-26 2015-09-08 Mountain View Pharmaceuticals, Inc. Polymer conjugates of interferon-beta with enhanced biological potency
US8247381B2 (en) 2003-03-14 2012-08-21 Biogenerix Ag Branched water-soluble polymers and their conjugates
US7803777B2 (en) 2003-03-14 2010-09-28 Biogenerix Ag Branched water-soluble polymers and their conjugates
US8853161B2 (en) 2003-04-09 2014-10-07 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
US8063015B2 (en) 2003-04-09 2011-11-22 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
US9005625B2 (en) 2003-07-25 2015-04-14 Novo Nordisk A/S Antibody toxin conjugates
US8916360B2 (en) 2003-11-24 2014-12-23 Novo Nordisk A/S Glycopegylated erythropoietin
US8633157B2 (en) 2003-11-24 2014-01-21 Novo Nordisk A/S Glycopegylated erythropoietin
US7842661B2 (en) 2003-11-24 2010-11-30 Novo Nordisk A/S Glycopegylated erythropoietin formulations
US7956032B2 (en) 2003-12-03 2011-06-07 Novo Nordisk A/S Glycopegylated granulocyte colony stimulating factor
US8361961B2 (en) 2004-01-08 2013-01-29 Biogenerix Ag O-linked glycosylation of peptides
US8791066B2 (en) 2004-07-13 2014-07-29 Novo Nordisk A/S Branched PEG remodeling and glycosylation of glucagon-like peptide-1 [GLP-1]
US8268967B2 (en) 2004-09-10 2012-09-18 Novo Nordisk A/S Glycopegylated interferon α
US9200049B2 (en) 2004-10-29 2015-12-01 Novo Nordisk A/S Remodeling and glycopegylation of fibroblast growth factor (FGF)
US10874714B2 (en) 2004-10-29 2020-12-29 89Bio Ltd. Method of treating fibroblast growth factor 21 (FGF-21) deficiency
US9029331B2 (en) 2005-01-10 2015-05-12 Novo Nordisk A/S Glycopegylated granulocyte colony stimulating factor
US9187546B2 (en) 2005-04-08 2015-11-17 Novo Nordisk A/S Compositions and methods for the preparation of protease resistant human growth hormone glycosylation mutants
US8911967B2 (en) 2005-08-19 2014-12-16 Novo Nordisk A/S One pot desialylation and glycopegylation of therapeutic peptides
US8841439B2 (en) 2005-11-03 2014-09-23 Novo Nordisk A/S Nucleotide sugar purification using membranes
US9187532B2 (en) 2006-07-21 2015-11-17 Novo Nordisk A/S Glycosylation of peptides via O-linked glycosylation sequences
US7893019B2 (en) 2006-08-11 2011-02-22 Bio-Ker S.R.L. G-CSF site-specific mono-conjugates
US8969532B2 (en) 2006-10-03 2015-03-03 Novo Nordisk A/S Methods for the purification of polypeptide conjugates comprising polyalkylene oxide using hydrophobic interaction chromatography
US9050304B2 (en) 2007-04-03 2015-06-09 Ratiopharm Gmbh Methods of treatment using glycopegylated G-CSF
US9493499B2 (en) 2007-06-12 2016-11-15 Novo Nordisk A/S Process for the production of purified cytidinemonophosphate-sialic acid-polyalkylene oxide (CMP-SA-PEG) as modified nucleotide sugars via anion exchange chromatography
US8207112B2 (en) 2007-08-29 2012-06-26 Biogenerix Ag Liquid formulation of G-CSF conjugate
US9150848B2 (en) 2008-02-27 2015-10-06 Novo Nordisk A/S Conjugated factor VIII molecules

Also Published As

Publication number Publication date
US4847325A (en) 1989-07-11
DE68913520T2 (en) 1994-08-25
IL89001A0 (en) 1989-08-15
AU3180789A (en) 1989-08-11
CA1340297C (en) 1998-12-29
EP0402378A1 (en) 1990-12-19
IL89001A (en) 1994-04-12
JPH03503759A (en) 1991-08-22
EP0402378B1 (en) 1994-03-02
IE890116L (en) 1989-07-20
DE68913520D1 (en) 1994-04-07
IE64887B1 (en) 1995-09-20
JP3280016B2 (en) 2002-04-30

Similar Documents

Publication Publication Date Title
US5153265A (en) Conjugation of polymer to colony stimulating factor-1
EP0402378B1 (en) Conjugation of polymer to colony stimulating factor-1
US4766106A (en) Solubilization of proteins for pharmaceutical compositions using polymer conjugation
US4917888A (en) Solubilization of immunotoxins for pharmaceutical compositions using polymer conjugation
EP0229108B1 (en) Solubilization of proteins for pharmaceutical compositions using polymer conjugation
US7090835B2 (en) N-terminally chemically modified protein compositions and methods
JP4157277B2 (en) Substantially pure histidine binding protein-polymer conjugate
EP0247860B1 (en) Tumor necrosis factor formulation and its preparation
US20030190304A1 (en) Pegylation reagents and compounds formed therewith
AU2006278490A1 (en) Conjugates of a G-CSF moiety and a polymer
US20030053982A1 (en) N-terminally chemically modified protein compositions and methods
AU580431B2 (en) Solubilization of proteins for pharmaceutical compositions using polymer conjugation
PT89861B (en) PROCESS FOR THE CONJUGATION OF POLYMERES WITH COLONIAL STIMULATOR FACTOR-1
Gabriel et al. United States Patent (19)
AU2004235682B2 (en) N-Terminally chemically modified protein compositions and methods

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU BG BR DK FI HU JP KR LK NO RO SU US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE FR GB IT LU NL SE

WWE Wipo information: entry into national phase

Ref document number: 1989902670

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1989902670

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1989902670

Country of ref document: EP